1
|
Santos VC, Wijesekera N, El-Mayet FS, Jones C. Glucocorticoid receptor and specificity protein 1 (Sp1) or Sp3 transactivate HSV-1 ICP0 promoter sequences but a GC-rich binding antibiotic, Mithramycin A, impairs reactivation from latency. Virus Res 2024; 350:199487. [PMID: 39490590 PMCID: PMC11570323 DOI: 10.1016/j.virusres.2024.199487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 10/07/2024] [Accepted: 10/17/2024] [Indexed: 11/05/2024]
Abstract
Glucocorticoid receptor (GR) activation enhances Human alpha-herpes virus 1 (HSV-1) replication and explant-induced reactivation from latency. Furthermore, GR and Krüppel-like factor 15 (KLF15) cooperatively transactivate cis-regulatory modules (CRMs) that drive expression of infected cell protein 0 (ICP0), ICP4, and ICP27. KLF and specificity protein (Sp) family members bind GC-rich or C-rich sequences and belong to the same super-family of transcription factors. Based on these observations, we hypothesized CRMs spanning the ICP0 promoter are transactivated by GR and Sp1 or Sp3. CRM-A (-800 to -635), CRM-B (-485 to -635), and CRM-D (-232 to -24), but not CRM-C, were significantly transactivated by GR, DEX, and Sp1 or Sp3 in mouse neuroblastoma cells (Neuro-2A). Mutagenesis of Sp1/Sp3 binding sites were important for transactivation of CRM-A and CRM-B. Chromatin immunoprecipitation studies revealed significantly higher levels of GR occupied ICP0 promoter sequences when Sp1 or Sp3 was over-expressed suggesting these transcriptions factors recruit GR to ICP0 CRM sequences. Mithramycin A, an antibiotic that preferentially binds GC-rich DNA and impairs Sp1/Sp3 dependent transactivation and reduced virus shedding during reactivation from latency in mice latently infected with HSV-1. These studies indicate GR and certain stress-induced cellular transcription factors preferentially bind GC rich DNA, which stimulates HSV-1 gene expression and reactivation from latency in trigeminal ganglia of latently infected mice.
Collapse
Affiliation(s)
- Vanessa Claire Santos
- Oklahoma State University, College of Veterinary Medicine, Department of Veterinary Pathobiology, Stillwater, OK 74078, USA.
| | - Nishani Wijesekera
- Oklahoma State University, College of Veterinary Medicine, Department of Veterinary Pathobiology, Stillwater, OK 74078, USA.
| | - Fouad S El-Mayet
- Oklahoma State University, College of Veterinary Medicine, Department of Veterinary Pathobiology, Stillwater, OK 74078, USA; Benha University, Faculty of Veterinary Medicine, Department of Virology, Benha, Egypt.
| | - Clinton Jones
- Oklahoma State University, College of Veterinary Medicine, Department of Veterinary Pathobiology, Stillwater, OK 74078, USA.
| |
Collapse
|
2
|
El-Mayet FS, Santos VC, Wijesekera N, Lubbers S, Harrison KS, Sadeghi H, Jones C. Glucocorticoid receptor and specificity protein 1 (Sp1) or Sp3, but not the antibiotic Mithramycin A, stimulates human alphaherpesvirus 1 (HSV-1) replication. Antiviral Res 2024; 225:105870. [PMID: 38556059 PMCID: PMC11109923 DOI: 10.1016/j.antiviral.2024.105870] [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] [Received: 02/04/2024] [Revised: 03/15/2024] [Accepted: 03/22/2024] [Indexed: 04/02/2024]
Abstract
Following acute human alphaherpesvirus 1 (HSV-1) infection of oral-facial mucosal surfaces, sensory neurons in trigeminal ganglia (TG) are important sites for life-long latency. Neurons in the central nervous system, including brainstem, also harbor viral genomes during latency. Periodically, certain cellular stressors trigger reactivation from latency, which can lead to recurrent HSV-1 disease: herpes labialis, herpes stromal keratitis, and encephalitis for example. Activation of the glucocorticoid receptor (GR) by stressful stimuli enhances HSV-1 gene expression, replication, and explant-induced reactivation. GR and certain stress-induced Krüppel like factors (KLF) cooperatively transactivate cis-regulatory modules (CRM) that drive expression of viral transcriptional regulatory proteins (ICP0, ICP4, and ICP27). These CRMs lack GR response elements (GRE); however, specificity protein 1 (Sp1) binding sites are crucial for GR and KLF15 or KLF4 mediated transactivation. Hence, we tested whether Sp1 or Sp3 regulate viral replication and transactivation of the ICP0 promoter. During early stages of explant-induced reactivation from latency, the number of Sp3+ TG neurons were significantly higher relative to TG from latently infected mice. Conversely, Sp1+ TG neurons were only increased in females, but not male mice, during explant-induced reactivation. Sp1 siRNA significantly reduced HSV-1 replication in cultured mouse (Neuro-2A) and monkey (CV-1) cells. Mithramycin A, an antibiotic that has anti-tumor activity preferentially interacts with GC-rich DNA, including Sp1 binding sites, significantly reduced HSV-1 replication indicating it has antiviral activity. GR and Sp1 or Sp3 transactivated the HSV-1 ICP0 promoter in Neuro-2A and CV-1 cells confirming these transcription factors enhance viral replication and gene expression.
Collapse
Affiliation(s)
- Fouad S El-Mayet
- Oklahoma State University, College of Veterinary Medicine, Department of Veterinary Pathobiology, Stillwater, OK, USA; Benha University, Faculty of Veterinary Medicine, Department of Virology, Moshtohor, 13736, Kaliobyia, Egypt
| | - Vanessa Claire Santos
- Oklahoma State University, College of Veterinary Medicine, Department of Veterinary Pathobiology, Stillwater, OK, USA
| | - Nishani Wijesekera
- Oklahoma State University, College of Veterinary Medicine, Department of Veterinary Pathobiology, Stillwater, OK, USA
| | - Sydney Lubbers
- Oklahoma State University, College of Veterinary Medicine, Department of Veterinary Pathobiology, Stillwater, OK, USA
| | - Kelly S Harrison
- Oklahoma State University, College of Veterinary Medicine, Department of Veterinary Pathobiology, Stillwater, OK, USA
| | - Hafez Sadeghi
- Oklahoma State University, College of Veterinary Medicine, Department of Veterinary Pathobiology, Stillwater, OK, USA
| | - Clinton Jones
- Oklahoma State University, College of Veterinary Medicine, Department of Veterinary Pathobiology, Stillwater, OK, USA.
| |
Collapse
|
3
|
Li R, Song B, Xu L, Zheng J, Pan W, Cai F, Wang J, Wu Y, Song W. Regulation of USP25 by SP1 Associates with Amyloidogenesis. J Alzheimers Dis 2023; 92:1459-1472. [PMID: 36938736 DOI: 10.3233/jad-221184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/15/2023]
Abstract
BACKGROUND Trisomy 21, an extra copy of human chromosome 21 (HSA21), causes most Down's syndrome (DS) cases. Individuals with DS inevitably develop Alzheimer's disease (AD) neuropathological phenotypes after middle age including amyloid plaques and tau neurofibrillary tangles. Ubiquitin Specific Peptidase 25 (USP25), encoding by USP25 gene located on HSA21, is a deubiquitinating enzyme, which plays an important role in both DS and AD pathogenesis. However, the regulation of USP25 remains unclear. OBJECTIVE We aimed to determine the regulation of USP25 by specificity protein 1 (SP1) in neuronal cells and its potential role in amyloidogenesis. METHODS The transcription start site and promoter activity was identified by SMART-RACE and Dual-luciferase assay. Functional SP1-responsive elements were examined by EMSA. USP25 expression was examined by RT-PCR and immunoblotting. Student's t-test or one-way ANOVA were applied or statistical analysis. RESULTS The transcription start site of human USP25 gene was identified. Three functional SP1 responsive elements in human USP25 gene were revealed. SP1 promotes USP25 transcription and subsequent USP25 protein expression, while SP1 inhibition significantly reduces USP25 expression in both non-neuronal and neuronal cells. Moreover, SP1 inhibition dramatically reduces amyloidogenesis. CONCLUSION We demonstrates that transcription factor SP1 regulates USP25 gene expression, which associates with amyloidogenesis. It suggests that SP1 signaling may play an important role in USP25 regulation and contribute to USP25-mediated DS and AD pathogenesis.
Collapse
Affiliation(s)
- Ran Li
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China.,Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province,Zhejiang Provincial Clinical Research Center for Mental Disorders, School of Mental Health and Kangning Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Beibei Song
- Townsend Family Laboratories, Department of Psychiatry, Graduate Program in Neuroscience, The University of British Columbia, Vancouver, BC, Canada
| | - Lu Xu
- Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province,Zhejiang Provincial Clinical Research Center for Mental Disorders, School of Mental Health and Kangning Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jiali Zheng
- Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province,Zhejiang Provincial Clinical Research Center for Mental Disorders, School of Mental Health and Kangning Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Wenhao Pan
- Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province,Zhejiang Provincial Clinical Research Center for Mental Disorders, School of Mental Health and Kangning Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Fang Cai
- Townsend Family Laboratories, Department of Psychiatry, Graduate Program in Neuroscience, The University of British Columbia, Vancouver, BC, Canada
| | - Juelu Wang
- Townsend Family Laboratories, Department of Psychiatry, Graduate Program in Neuroscience, The University of British Columbia, Vancouver, BC, Canada
| | - Yili Wu
- Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province,Zhejiang Provincial Clinical Research Center for Mental Disorders, School of Mental Health and Kangning Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China.,Oujiang Laboratory Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Wenzhou, Zhejiang, China
| | - Weihong Song
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China.,Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province,Zhejiang Provincial Clinical Research Center for Mental Disorders, School of Mental Health and Kangning Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China.,Oujiang Laboratory Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Wenzhou, Zhejiang, China
| |
Collapse
|
4
|
Hay J, Tarafdar A, Holroyd AK, Moka HA, Dunn KM, Alshayeb A, Lloyd BH, Cassels J, Malik N, Khan AF, Sou I, Lees J, Almuhanna HNB, Kalakonda N, Slupsky JR, Michie AM. PKCβ Facilitates Leukemogenesis in Chronic Lymphocytic Leukaemia by Promoting Constitutive BCR-Mediated Signalling. Cancers (Basel) 2022; 14:cancers14236006. [PMID: 36497487 PMCID: PMC9735720 DOI: 10.3390/cancers14236006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 11/22/2022] [Accepted: 11/30/2022] [Indexed: 12/13/2022] Open
Abstract
B cell antigen receptor (BCR) signalling competence is critical for the pathogenesis of chronic lymphocytic leukaemia (CLL). Defining key proteins that facilitate these networks aid in the identification of targets for therapeutic exploitation. We previously demonstrated that reduced PKCα function in mouse hematopoietic stem/progenitor cells (HPSCs) resulted in PKCβII upregulation and generation of a poor-prognostic CLL-like disease. Here, prkcb knockdown in HSPCs leads to reduced survival of PKCα-KR-expressing CLL-like cells, concurrent with reduced expression of the leukemic markers CD5 and CD23. SP1 promotes elevated expression of prkcb in PKCα-KR expressing cells enabling leukemogenesis. Global gene analysis revealed an upregulation of genes associated with B cell activation in PKCα-KR expressing cells, coincident with upregulation of PKCβII: supported by activation of key signalling hubs proximal to the BCR and elevated proliferation. Ibrutinib (BTK inhibitor) or enzastaurin (PKCβII inhibitor) treatment of PKCα-KR expressing cells and primary CLL cells showed similar patterns of Akt/mTOR pathway inhibition, supporting the role for PKCβII in maintaining proliferative signals in our CLL mouse model. Ibrutinib or enzastaurin treatment also reduced PKCα-KR-CLL cell migration towards CXCL12. Overall, we demonstrate that PKCβ expression facilitates leukemogenesis and identify that BCR-mediated signalling is a key driver of CLL development in the PKCα-KR model.
Collapse
Affiliation(s)
- Jodie Hay
- School of Cancer Sciences, College of Medicine, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
- Paul O’Gorman Leukaemia Research Centre, Gartnavel General Hospital, 21 Shelley Road, Glasgow G12 0ZD, UK
| | - Anuradha Tarafdar
- School of Cancer Sciences, College of Medicine, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Ailsa K. Holroyd
- School of Cancer Sciences, College of Medicine, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Hothri A. Moka
- School of Cancer Sciences, College of Medicine, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Karen M. Dunn
- School of Cancer Sciences, College of Medicine, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
- Paul O’Gorman Leukaemia Research Centre, Gartnavel General Hospital, 21 Shelley Road, Glasgow G12 0ZD, UK
| | - Alzahra Alshayeb
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7BE, UK
| | - Bryony H. Lloyd
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7BE, UK
| | - Jennifer Cassels
- School of Cancer Sciences, College of Medicine, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
- Paul O’Gorman Leukaemia Research Centre, Gartnavel General Hospital, 21 Shelley Road, Glasgow G12 0ZD, UK
| | - Natasha Malik
- School of Cancer Sciences, College of Medicine, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Ashfia F. Khan
- School of Cancer Sciences, College of Medicine, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - IengFong Sou
- School of Cancer Sciences, College of Medicine, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Jamie Lees
- School of Cancer Sciences, College of Medicine, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
- Paul O’Gorman Leukaemia Research Centre, Gartnavel General Hospital, 21 Shelley Road, Glasgow G12 0ZD, UK
| | - Hassan N. B. Almuhanna
- School of Cancer Sciences, College of Medicine, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
- Paul O’Gorman Leukaemia Research Centre, Gartnavel General Hospital, 21 Shelley Road, Glasgow G12 0ZD, UK
| | - Nagesh Kalakonda
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7BE, UK
| | - Joseph R. Slupsky
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7BE, UK
| | - Alison M. Michie
- School of Cancer Sciences, College of Medicine, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
- Paul O’Gorman Leukaemia Research Centre, Gartnavel General Hospital, 21 Shelley Road, Glasgow G12 0ZD, UK
- Correspondence: ; Tel.: +44-(0)141-301-7885
| |
Collapse
|
5
|
Isagulieva AK, Tevyashova AN, Shtil AA. Aureolic Acid-Derived Antibiotics: Prospects for a Biologically Active Class. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2022. [DOI: 10.1134/s1068162022020121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
6
|
Khader N, Shchuka VM, Shynlova O, Mitchell JA. Transcriptional control of parturition: insights from gene regulation studies in the myometrium. Mol Hum Reprod 2021; 27:gaab024. [PMID: 33823545 PMCID: PMC8126590 DOI: 10.1093/molehr/gaab024] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 03/09/2021] [Indexed: 12/19/2022] Open
Abstract
The onset of labour is a culmination of a series of highly coordinated and preparatory physiological events that take place throughout the gestational period. In order to produce the associated contractions needed for foetal delivery, smooth muscle cells in the muscular layer of the uterus (i.e. myometrium) undergo a transition from quiescent to contractile phenotypes. Here, we present the current understanding of the roles transcription factors play in critical labour-associated gene expression changes as part of the molecular mechanistic basis for this transition. Consideration is given to both transcription factors that have been well-studied in a myometrial context, i.e. activator protein 1, progesterone receptors, oestrogen receptors, and nuclear factor kappa B, as well as additional transcription factors whose gestational event-driving contributions have been demonstrated more recently. These transcription factors may form pregnancy- and labour-associated transcriptional regulatory networks in the myometrium to modulate the timing of labour onset. A more thorough understanding of the transcription factor-mediated, labour-promoting regulatory pathways holds promise for the development of new therapeutic treatments that can be used for the prevention of preterm labour in at-risk women.
Collapse
Affiliation(s)
- Nawrah Khader
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | - Virlana M Shchuka
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | - Oksana Shynlova
- Lunenfeld Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
- Department of Obstetrics & Gynaecology, University of Toronto, ON, Canada
| | - Jennifer A Mitchell
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
7
|
Yan X, Li M, Luo Z, Zhao Y, Zhang H, Chen L. VIP Induces Changes in the F-/G-Actin Ratio of Schlemm's Canal Endothelium via LRRK2 Transcriptional Regulation. Invest Ophthalmol Vis Sci 2021; 61:45. [PMID: 32572455 PMCID: PMC7415318 DOI: 10.1167/iovs.61.6.45] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose A previous study reported that vasoactive intestinal peptide (VIP) can regulate the cytoskeleton of Schlemm's canal (SC) endothelium and expand the SC lumen in a rat glaucoma model. In this study, we aimed to investigate the molecular mechanism of VIP on cytoskeleton regulation. Methods During in vivo experiments in rats, leucine-rich repeat kinase 2 (LRRK2) expression and the ratio of F-actin to G-actin (F-/G-actin) surrounding SC were examined by immunofluorescence after the application of VIP. For in vitro experiments in human umbilical vein endothelial cells, both quantitative PCR (qPCR) and western blotting were performed to evaluate Sp1 and LRRK2 expression after the application of VIP (and Sp1/LRRK2 inhibitor). In addition, the F-/G-actin ratio was examined by both immunofluorescence and western blotting after the application of VIP (and LRRK2 inhibitor). Results VIP induced increases in the expression of LRRK2 both in vivo and in vitro and the nuclear translocation of Sp1 in vitro. The application of Sp1 inhibitor abolished the increase in LRRK2 expression induced by VIP in vitro. In addition, VIP changed the F-/G-actin ratio, and this effect was abolished by the LRRK2 inhibitor both in vivo and in vitro. Conclusions VIP increased the expression of LRRK2, and this regulation was due to the nuclear translocation of Sp1. VIP further changed the F-/G-actin ratio and regulated the balance between the stabilization and destabilization of the F-actin architecture. This study elucidates a novel mechanism by which VIP regulates the actin cytoskeleton of SC endothelium via the Sp1–LRRK2 pathway, suggesting a potential novel treatment strategy for glaucoma.
Collapse
|
8
|
Lang C, Dai Y, Wu Z, Yang Q, He S, Zhang X, Guo W, Lai Y, Du H, Wang H, Ren D, Peng X. SMAD3/SP1 complex-mediated constitutive active loop between lncRNA PCAT7 and TGF-β signaling promotes prostate cancer bone metastasis. Mol Oncol 2020; 14:808-828. [PMID: 31925912 PMCID: PMC7138406 DOI: 10.1002/1878-0261.12634] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 12/16/2019] [Accepted: 01/09/2020] [Indexed: 01/05/2023] Open
Abstract
Bone metastasis is associated with cancer-related death in patients with prostate cancer (PCa). Long noncoding RNAs (lncRNAs) play critical roles in tumor progression of PCa. Nevertheless, the biological function of lncRNAs in PCa bone metastasis remains unclear. PCAT7 was identified as a bone metastasis-related lncRNA via analyzing TCGA dataset. Meanwhile, PCAT7 was found to be elevated in primary PCa tissues with bone metastasis and associated with bone metastasis status and poor prognosis of patients with PCa. Functionally, our results reveal that PCAT7 overexpression promotes PCa bone metastasis in vivo, as well as migration, invasion, and EMT of PCa cells in vitro; on the contrary, PCAT7 knockdown has an inverse effect. Mechanistically, PCAT7 activates TGF-β/SMAD signaling by upregulating TGFBR1 expression via sponging miR-324-5p. In turn, TGF-β signaling forms a positive feedback loop with PCAT7 via SMAD3/SP1 complex-induced PCAT7 upregulation. Finally, the clinical positive correlation between PCAT7 and TGFBR1 and TGF-β signaling activity, and the negative association with miR-324-5p are further demonstrated in PCa tissues and clinical primary PCa cells. This study reveals a novel mechanism that is responsible for the constitutive activation of TGF-β signaling in PCa bone metastasis, implying that PCAT7 can act as a potential therapeutic target against bone metastasis of PCa via disrupting the constitutive active loop between PCAT7 and TGF-β signaling.
Collapse
Affiliation(s)
- Chuandong Lang
- Department of Orthopaedic SurgeryThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Orthopedics and TraumatologyGuangzhouChina
| | - Yuhu Dai
- Department of Orthopaedic SurgeryThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Orthopedics and TraumatologyGuangzhouChina
| | - Zhengquan Wu
- Department of Orthopaedic SurgeryThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Orthopedics and TraumatologyGuangzhouChina
| | - Qing Yang
- Department of Orthopaedic SurgeryThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Orthopedics and TraumatologyGuangzhouChina
| | - Shaofu He
- Department of RadiologyThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Xin Zhang
- Clinical Experimental CenterJiangmen Central HospitalAffiliated Jiangmen HospitalSun Yat‐sen UniversityJiangmenChina
| | - Wei Guo
- Department of Orthopaedic SurgeryThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Orthopedics and TraumatologyGuangzhouChina
| | - Yingrong Lai
- Department of PathologyThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Hong Du
- Department of PathologyThe First People’s Hospital of Guangzhou CityGuangzhouChina
| | - Hehe Wang
- Department of Medical LaboratoryWeifang Medical UniversityWeifangChina
| | - Dong Ren
- Department of Orthopaedic SurgeryThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Orthopedics and TraumatologyGuangzhouChina
| | - Xinsheng Peng
- Department of Orthopaedic SurgeryThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Orthopedics and TraumatologyGuangzhouChina
| |
Collapse
|
9
|
Screening Method for Anti-Colon Cancer Drugs Using Two Sensor Cell Lines with Human β4-Galactosyltransferase 4 Gene Promoters. SENSORS 2018; 18:s18082573. [PMID: 30082623 PMCID: PMC6111286 DOI: 10.3390/s18082573] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 07/27/2018] [Accepted: 08/05/2018] [Indexed: 12/29/2022]
Abstract
The increased expression of β4-galactosyltransferase (β4GalT) 4 is closely associated with poor prognosis of colon cancer. Recently, we showed that the expression of the β4GalT4 gene is regulated by the 0.17 kb core promoter region containing one binding site for Specificity protein 1 (Sp1). To develop a screening method for anti-colon cancer drugs, two sensor cell lines having the luciferase gene under the control of two β4GalT4 gene promoters that differed in length were established from SW480 human colon cancer cells. The hGT4-0.17-sensor cells possessed the luciferase reporter driven by the 0.17 kb promoter, while the hGT4-0.3-sensor cells possessed the luciferase reporter driven by the 0.3 kb promoter containing one binding site each for colon cancer-related transcription factors including activator protein 2, E2F, caudal-related homeobox transcription factors, and Runt-related transcription factors besides Sp1. Upon treatment with mitogen-activated protein kinase signaling inhibitor U0126, the promoter activities of the hGT4-0.3-sensor cells decreased significantly, while those of the hGT4-0.17-sensor cells remained unchanged. These results suggest that the responsiveness to U0126 differs between two sensor cell lines due to the different regulation of the luciferase reporters. This study provides the screening method for anti-colon cancer drugs by the combination of two sensor cell lines.
Collapse
|
10
|
Lambert M, Jambon S, Depauw S, David-Cordonnier MH. Targeting Transcription Factors for Cancer Treatment. Molecules 2018; 23:molecules23061479. [PMID: 29921764 PMCID: PMC6100431 DOI: 10.3390/molecules23061479] [Citation(s) in RCA: 241] [Impact Index Per Article: 34.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 06/11/2018] [Accepted: 06/15/2018] [Indexed: 12/15/2022] Open
Abstract
Transcription factors are involved in a large number of human diseases such as cancers for which they account for about 20% of all oncogenes identified so far. For long time, with the exception of ligand-inducible nuclear receptors, transcription factors were considered as “undruggable” targets. Advances knowledge of these transcription factors, in terms of structure, function (expression, degradation, interaction with co-factors and other proteins) and the dynamics of their mode of binding to DNA has changed this postulate and paved the way for new therapies targeted against transcription factors. Here, we discuss various ways to target transcription factors in cancer models: by modulating their expression or degradation, by blocking protein/protein interactions, by targeting the transcription factor itself to prevent its DNA binding either through a binding pocket or at the DNA-interacting site, some of these inhibitors being currently used or evaluated for cancer treatment. Such different targeting of transcription factors by small molecules is facilitated by modern chemistry developing a wide variety of original molecules designed to specifically abort transcription factor and by an increased knowledge of their pathological implication through the use of new technologies in order to make it possible to improve therapeutic control of transcription factor oncogenic functions.
Collapse
Affiliation(s)
- Mélanie Lambert
- INSERM UMR-S1172-JPARC (Jean-Pierre Aubert Research Center), Lille University and Hospital Center (CHU-Lille), Institut pour la Recherche sur le Cancer de Lille (IRCL), Place de Verdun, F-59045 Lille, France.
| | - Samy Jambon
- INSERM UMR-S1172-JPARC (Jean-Pierre Aubert Research Center), Lille University and Hospital Center (CHU-Lille), Institut pour la Recherche sur le Cancer de Lille (IRCL), Place de Verdun, F-59045 Lille, France.
| | - Sabine Depauw
- INSERM UMR-S1172-JPARC (Jean-Pierre Aubert Research Center), Lille University and Hospital Center (CHU-Lille), Institut pour la Recherche sur le Cancer de Lille (IRCL), Place de Verdun, F-59045 Lille, France.
| | - Marie-Hélène David-Cordonnier
- INSERM UMR-S1172-JPARC (Jean-Pierre Aubert Research Center), Lille University and Hospital Center (CHU-Lille), Institut pour la Recherche sur le Cancer de Lille (IRCL), Place de Verdun, F-59045 Lille, France.
| |
Collapse
|
11
|
Choi MC, Choi WH. Mithramycin A Alleviates Osteoarthritic Cartilage Destruction by Inhibiting HIF-2α Expression. Int J Mol Sci 2018; 19:ijms19051411. [PMID: 29747385 PMCID: PMC5983647 DOI: 10.3390/ijms19051411] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 04/25/2018] [Accepted: 05/04/2018] [Indexed: 11/16/2022] Open
Abstract
Osteoarthritis (OA) is the most common and increasing joint disease worldwide. Current treatment for OA is limited to control of symptoms. The purpose of this study was to determine the effect of specificity protein 1 (SP1) inhibitor Mithramycin A (MitA) on chondrocyte catabolism and OA pathogenesis and to explore the underlying molecular mechanisms involving SP1 and other key factors that are critical for OA. Here, we show that MitA markedly inhibited expressions of matrix-degrading enzymes induced by pro-inflammatory cytokine interleukin-1β (IL-1β) in mouse primary chondrocytes. Intra-articular injection of MitA into mouse knee joint alleviated OA cartilage destruction induced by surgical destabilization of the medial meniscus (DMM). However, modulation of SP1 level in chondrocyte and mouse cartilage did not alter catabolic gene expression or cartilage integrity, respectively. Instead, MitA significantly impaired the expression of HIF-2α known to be critical for OA pathogenesis. Such reduction in expression of HIF-2α by MitA was caused by inhibition of NF-κB activation, at least in part. These results suggest that MitA can alleviate OA pathogenesis by suppressing NF-κB-HIF-2α pathway, thus providing insight into therapeutic strategy for OA.
Collapse
Affiliation(s)
- Moon-Chang Choi
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, Korea.
- Department of Biomedical Science, Chosun University, Gwangju 61452, Korea.
| | - Woo Hee Choi
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, Korea.
| |
Collapse
|
12
|
Niu C, Li M, Zhu S, Chen Y, Zhou L, Xu D, Li W, Cui J, Liu Y, Chen J. Decitabine Inhibits Gamma Delta T Cell Cytotoxicity by Promoting KIR2DL2/3 Expression. Front Immunol 2018; 9:617. [PMID: 29632540 PMCID: PMC5879086 DOI: 10.3389/fimmu.2018.00617] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 03/12/2018] [Indexed: 12/25/2022] Open
Abstract
Gamma delta (γδ) T cells, which possess potent cytotoxicity against a wide range of cancer cells, have become a potential avenue for adoptive immunotherapy. Decitabine (DAC) has been reported to enhance the immunogenicity of tumor cells, thereby reinstating endogenous immune recognition and tumor lysis. However, DAC has also been demonstrated to have direct effects on immune cells. In this study, we report that DAC inhibits γδ T cell proliferation. In addition, DAC increases the number of KIR2DL2/3-positive γδ T cells, which are less cytotoxic than the KIR2DL2/3-negative γδ T cells. We found that DAC upregulated KIR2DL2/3 expression in KIR2DL2/3-negative γδ T cells by inhibiting KIR2DL2/3 promoter methylation, which enhances the binding of KIR2DL2/3 promoter to Sp-1 and activates KIR2DL2/3 gene expression. Our data demonstrated that DAC can inhibit the function of human γδ T cells at both cellular and molecular levels, which confirms and extrapolates the results of previous studies showing that DAC can negatively regulate the function of NK cells and αβ T cells of the immune system.
Collapse
Affiliation(s)
- Chao Niu
- Department of Translational Medicine, The First Hospital of Jilin University, Changchun, China.,Department of Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Min Li
- Department of Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Shan Zhu
- Department of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | - Yongchong Chen
- Department of Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Lei Zhou
- Department of Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Dongsheng Xu
- Department of Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Wei Li
- Department of Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Jiuwei Cui
- Department of Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Yongjun Liu
- Department of Translational Medicine, The First Hospital of Jilin University, Changchun, China.,Sanofi Research and Development, Cambridge, MA, United States
| | - Jingtao Chen
- Department of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
13
|
Muramoto K, Tange R, Ishii T, Miyauchi K, Sato T. Downregulation of Transcription Factor Sp1 Suppresses Malignant Properties of A549 Human Lung Cancer Cell Line with Decreased β4-Galactosylation of Highly Branched N-Glycans. Biol Pharm Bull 2017; 40:1282-1288. [PMID: 28529241 DOI: 10.1248/bpb.b17-00212] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2023]
Abstract
Dramatic changes in the glycan structures of cell surface proteins have been observed upon malignant transformation of cells as induced by the altered expression levels of glycosyltransferases. Such changes are closely associated with the malignant properties of cancer cells. Transcription factor Sp1 regulates the gene expression of various molecules including glycosyltransferases. Herein, we investigated whether or not Sp1-downregulation affects to N-glycosylation of glycoproteins and malignant properties of A549 human lung cancer cell line. We established a stable clone whose Sp1-expression level was reduced to 50% of a control clone by RNA interference. Lectin blotting revealed that the β4-galactosylation of highly branched N-glycans decreases mainly in cell adhesion molecule, E-cadherin. The analysis of underlying mechanism for decreased β4-galactosylation of N-glycans showed that the gene expression level of β4-galactosyltransferase (β4GalT) 1 decreases dramatically by downregulation of Sp1 without changes in those of β4GalT2 and N-acetylglucosaminyltransferase V. Mutations in the Sp1-binding sites of the β4GalT1 gene promoter showed that the promoter activity decreases significantly, indicating that the gene expression is regulated by Sp1. These results indicate that the β4-galactosylation of highly branched N-glycans decreases by downregulation of Sp1 through the reduced expression of the β4GalT1 gene. Furthermore, the Sp1-downregulated cells showed the suppression of the anchorage-independent growth in soft agar and migratory activity when compared to the control cells. The present study demonstrates that downregulation of Sp1 suppresses the malignant properties of A549 cells through the decreased β4-galactosylation of highly branched N-glycans.
Collapse
Affiliation(s)
- Kodai Muramoto
- Laboratory of Glycobiology, Department of Bioengineering, Nagaoka University of Technology
| | - Riho Tange
- Laboratory of Glycobiology, Department of Bioengineering, Nagaoka University of Technology
| | - Takayuki Ishii
- Laboratory of Glycobiology, Department of Bioengineering, Nagaoka University of Technology
| | - Kana Miyauchi
- Laboratory of Glycobiology, Department of Bioengineering, Nagaoka University of Technology
| | - Takeshi Sato
- Laboratory of Glycobiology, Department of Bioengineering, Nagaoka University of Technology
| |
Collapse
|
14
|
Transcriptional mechanism of vascular endothelial growth factor-induced expression of protein kinase CβII in chronic lymphocytic leukaemia cells. Sci Rep 2017; 7:43228. [PMID: 28233872 PMCID: PMC5324130 DOI: 10.1038/srep43228] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 01/20/2017] [Indexed: 12/24/2022] Open
Abstract
A key feature of chronic lymphocytic leukaemia (CLL) cells is overexpressed protein kinase CβII (PKCβII), an S/T kinase important in the pathogenesis of this and other B cell malignancies. The mechanisms contributing to enhanced transcription of the gene coding for PKCβII, PRKCB, in CLL cells remain poorly described, but could be important because of potential insight into how the phenotype of these cells is regulated. Here, we show that SP1 is the major driver of PKCβII expression in CLL cells where enhanced association of this transcription factor with the PRKCB promoter is likely because of the presence of histone marks permissive of gene activation. We also show how vascular endothelial growth factor (VEGF) regulates PRKCB promoter function in CLL cells, stimulating PKCβ gene transcription via increased association of SP1 and decreased association of STAT3. Taken together, these results are the first to demonstrate a clear role for SP1 in the up regulation of PKCβII expression in CLL cells, and the first to link SP1 with the pathogenesis of this and potentially other B cell malignancies where PKCβII is overexpressed.
Collapse
|
15
|
Yu S, Yerges-Armstrong LM, Chu Y, Zmuda JM, Zhang Y. Transcriptional Regulation of Frizzled-1 in Human Osteoblasts by Sp1. PLoS One 2016; 11:e0163277. [PMID: 27695039 PMCID: PMC5047477 DOI: 10.1371/journal.pone.0163277] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 09/05/2016] [Indexed: 02/03/2023] Open
Abstract
The wingless pathway has a powerful influence on bone metabolism and is a therapeutic target in skeletal disorders. Wingless signaling is mediated in part through the Frizzled (FZD) receptor family. FZD transcriptional regulation is poorly understood. Herein we tested the hypothesis that Sp1 plays an important role in the transcriptional regulation of FZD1 expression in osteoblasts and osteoblast mineralization. To test this hypothesis, we conducted FZD1 promoter assays in Saos2 cells with and without Sp1 overexpression. We found that Sp1 significantly up-regulates FZD1 promoter activity in Saos2 cells. Chromatin immunoprecipitation (ChIP) and electrophoretic mobility shift (EMSA) assays identified a novel and functional Sp1 binding site at -44 to -40 from the translation start site in the FZD1 promoter. The Sp1-dependent activation of the FZD1 promoter was abolished by mithramycin A (MMA), an antibiotic affecting both Sp1 binding and Sp1 protein levels in Saos2 cells. Similarly, down-regulation of Sp1 in hFOB cells resulted in less FZD1 expression and lower alkaline phosphatase activity. Moreover, over-expression of Sp1 increased FZD1 expression and Saos2 cell mineralization while MMA decreased Sp1 and FZD1 expression and Saos2 cell mineralization. Knockdown of FZD1 prior to Sp1 overexpression partially abolished Sp1 stimulation of osteoblast differentiation markers. Taken together, our results suggest that Sp1 plays a role in human osteoblast differentiation and mineralization, which is at least partially mediated by Sp1-dependent transactivation of FZD1.
Collapse
Affiliation(s)
- Shibing Yu
- Department of Medicine, School of Medicine, University of Pittsburgh, Pennsylvania, United States of America
| | - Laura M. Yerges-Armstrong
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pennsylvania, United States of America
- Program in Personalized and Genomic Medicine and Department of Medicine, Division of Endocrinology, Diabetes and Nutrition, School of Medicine, University of Maryland, College Park, Maryland, United States of America
| | - Yanxia Chu
- Department of Medicine, School of Medicine, University of Pittsburgh, Pennsylvania, United States of America
| | - Joseph M. Zmuda
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pennsylvania, United States of America
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pennsylvania, United States of America
| | - Yingze Zhang
- Department of Medicine, School of Medicine, University of Pittsburgh, Pennsylvania, United States of America
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
16
|
Yanuaryska RD, Miyoshi K, Adiningrat A, Horiguchi T, Tanimura A, Hagita H, Noma T. Sp6 regulation of Rock1 promoter activity in dental epithelial cells. THE JOURNAL OF MEDICAL INVESTIGATION 2016; 61:306-17. [PMID: 25264049 DOI: 10.2152/jmi.61.306] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Sp6 is a transcription factor of the SP/KLF family and an indispensable regulator of the morphological dynamics of ameloblast differentiation during tooth development. However, the underlying molecular mechanisms remain unclear. We have previously identified one of the Sp6 downstream genes, Rock1, which is involved in ameloblast polarization. In this study, we investigated the transcriptional regulatory mechanisms of Rock1 by Sp6. First, we identified the transcription start sites (TSS) and cloned the 5'-flanking region of Rock1. Serial deletion analyses identified a critical region for Rock1 promoter activity within the 249-bp upstream region of TSS, and chromatin immunoprecipitation assays revealed Sp6-binding to this region. Subsequent transient transfection experiments showed that Rock1 promoter activity is enhanced by Sp6, but reduced by Sp1. Treatment of dental epithelial cells with the GC-selective DNA binding inhibitor, mithramycin A, affected Rock1 promoter activity in loss of enhancement by Sp6, but not repression by Sp1. Further site-directed mutagenesis indicated that the region from -206 to -150 contains responsive elements for Sp6. Taken together, we conclude that Sp6 positively regulates Rock1 transcription by direct binding to the Rock1 promoter region from -206 to -150, which functionally distinct from Sp1.
Collapse
Affiliation(s)
- Ryna Dwi Yanuaryska
- Department of Molecular Biology, Institute of Health Biosciences, the University of Tokushima Graduate School
| | | | | | | | | | | | | |
Collapse
|
17
|
Wang J, Song W. Regulation of LRRK2 promoter activity and gene expression by Sp1. Mol Brain 2016; 9:33. [PMID: 27004687 PMCID: PMC4802577 DOI: 10.1186/s13041-016-0215-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Accepted: 03/14/2016] [Indexed: 12/12/2022] Open
Abstract
Background The dopaminergic neurodegeneration in the nigrostriatal pathway is a prominent neuropathological feature of Parkinson’s disease (PD). Mutations in various genes have been linked to familial PD, and leucine-rich repeat kinase 2 (LRRK2) gene is one of them. LRRK2 is a large complex protein, belonging to the ROCO family of proteins. Recent studies suggest that the level of LRRK2 protein is one of the contributing factors to PD pathogenesis. However, it remains elusive how LRRK2 is regulated at the transcriptional and translational level. Results In this study, we cloned a 1738 bp 5’-flanking region of the human LRRK2 gene. The transcriptional start site (TSS) was located to 135 bp upstream of translational start site and the fragment −118 to +133 bp had the minimum promoter activity required for transcription. There were two functional Sp1- responsive elements on the human LRRK2 gene promoter revealed by electrophoretic mobility shift assay (EMSA). Sp1 overexpression promoted LRRK2 transcription and translation in the cellular model. On the contrary, application of mithramycin A inhibited LRRK2 transcriptional and translational activities. Conclusion This is the first study indicating that Sp1 signaling plays an important role in the regulation of human LRRK2 gene expression. It suggests that controlling LRRK2 level by manipulating Sp1 signaling may be beneficial to attenuate PD-related neuropathology.
Collapse
Affiliation(s)
- Juelu Wang
- Department of Psychiatry, Townsend Family Laboratories, Graduate Program in Neuroscience, The University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Weihong Song
- Department of Psychiatry, Townsend Family Laboratories, Graduate Program in Neuroscience, The University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada.
| |
Collapse
|
18
|
Jin H, Kanthasamy A, Harischandra DS, Kondru N, Ghosh A, Panicker N, Anantharam V, Rana A, Kanthasamy AG. Histone hyperacetylation up-regulates protein kinase Cδ in dopaminergic neurons to induce cell death: relevance to epigenetic mechanisms of neurodegeneration in Parkinson disease. J Biol Chem 2014; 289:34743-67. [PMID: 25342743 DOI: 10.1074/jbc.m114.576702] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The oxidative stress-sensitive protein kinase Cδ (PKCδ) has been implicated in dopaminergic neuronal cell death. However, little is known about the epigenetic mechanisms regulating PKCδ expression in neurons. Here, we report a novel mechanism by which the PKCδ gene can be regulated by histone acetylation. Treatment with histone deacetylase (HDAC) inhibitor sodium butyrate (NaBu) induced PKCδ expression in cultured neurons, brain slices, and animal models. Several other HDAC inhibitors also mimicked NaBu. The chromatin immunoprecipitation analysis revealed that hyperacetylation of histone H4 by NaBu is associated with the PKCδ promoter. Deletion analysis of the PKCδ promoter mapped the NaBu-responsive element to an 81-bp minimal promoter region. Detailed mutagenesis studies within this region revealed that four GC boxes conferred hyperacetylation-induced PKCδ promoter activation. Cotransfection experiments and Sp inhibitor studies demonstrated that Sp1, Sp3, and Sp4 regulated NaBu-induced PKCδ up-regulation. However, NaBu did not alter the DNA binding activities of Sp proteins or their expression. Interestingly, a one-hybrid analysis revealed that NaBu enhanced transcriptional activity of Sp1/Sp3. Overexpression of the p300/cAMP-response element-binding protein-binding protein (CBP) potentiated the NaBu-mediated transactivation potential of Sp1/Sp3, but expressing several HDACs attenuated this effect, suggesting that p300/CBP and HDACs act as coactivators or corepressors in histone acetylation-induced PKCδ up-regulation. Finally, using genetic and pharmacological approaches, we showed that NaBu up-regulation of PKCδ sensitizes neurons to cell death in a human dopaminergic cell model and brain slice cultures. Together, these results indicate that histone acetylation regulates PKCδ expression to augment nigrostriatal dopaminergic cell death, which could contribute to the progressive neuropathogenesis of Parkinson disease.
Collapse
Affiliation(s)
- Huajun Jin
- From the Department of Biomedical Sciences, Iowa Center for Advanced Neurotoxicology, Iowa State University, Ames, Iowa 50011
| | - Arthi Kanthasamy
- From the Department of Biomedical Sciences, Iowa Center for Advanced Neurotoxicology, Iowa State University, Ames, Iowa 50011
| | - Dilshan S Harischandra
- From the Department of Biomedical Sciences, Iowa Center for Advanced Neurotoxicology, Iowa State University, Ames, Iowa 50011
| | - Naveen Kondru
- From the Department of Biomedical Sciences, Iowa Center for Advanced Neurotoxicology, Iowa State University, Ames, Iowa 50011
| | - Anamitra Ghosh
- From the Department of Biomedical Sciences, Iowa Center for Advanced Neurotoxicology, Iowa State University, Ames, Iowa 50011
| | - Nikhil Panicker
- From the Department of Biomedical Sciences, Iowa Center for Advanced Neurotoxicology, Iowa State University, Ames, Iowa 50011
| | - Vellareddy Anantharam
- From the Department of Biomedical Sciences, Iowa Center for Advanced Neurotoxicology, Iowa State University, Ames, Iowa 50011
| | - Ajay Rana
- the Department of Molecular Pharmacology and Therapeutics, Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois 60153, and the Hines Veterans Affairs Medical Center, Hines, Illinois 60141
| | - Anumantha G Kanthasamy
- From the Department of Biomedical Sciences, Iowa Center for Advanced Neurotoxicology, Iowa State University, Ames, Iowa 50011,
| |
Collapse
|
19
|
Methamphetamine-induced toxicity: an updated review on issues related to hyperthermia. Pharmacol Ther 2014; 144:28-40. [PMID: 24836729 DOI: 10.1016/j.pharmthera.2014.05.001] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Accepted: 05/07/2014] [Indexed: 01/30/2023]
Abstract
Reports of methamphetamine-related emergency room visits suggest that elevated body temperature is a universal presenting symptom, with lethal overdoses generally associated with extreme hyperthermia. This review summarizes the available information on methamphetamine toxicity as it pertains to elevations in body temperature. First, a brief overview of thermoregulatory mechanisms is presented. Next, central and peripheral targets that have been considered for potential involvement in methamphetamine hyperthermia are discussed. Finally, future areas of investigation are proposed, as further studies are needed to provide greater insight into the mechanisms that mediate the alterations in body temperature elicited by methamphetamine.
Collapse
|
20
|
Aledo R, Costales P, Ciudad C, Noé V, Llorente-Cortes V, Badimon L. Molecular and functional characterization of LRP1 promoter polymorphism c.1-25 C>G (rs138854007). Atherosclerosis 2014; 233:178-85. [PMID: 24529141 DOI: 10.1016/j.atherosclerosis.2013.12.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Revised: 11/20/2013] [Accepted: 12/05/2013] [Indexed: 10/25/2022]
Abstract
The transcription of the Low-density lipoprotein receptor-related protein (LRP1) is upregulated by aggregated LDL (agLDL) and angiotensin II (AngII) in human vascular smooth muscle cells (hVSMC). The polymorphism c.1-25C>G creates a new GC-box in the LRP1 promoter recognized by Sp1/Sp3 transcription factors. The aims of this study were 1) to evaluate the impact of c.1-25C>G polymorphism on LRP1 transcriptional activity and expression, and 2) to examine the response of c.1-25C>G LRP1 promoter to LDL and AngII. EMSA and Luciferase assays in HeLa cells showed that -25G promoter has enhanced basal transcriptional activity and specific Sp1/Sp3 binding. hVSMC with GG genotype (GG-hVSMC) had higher LRP1 mRNA and protein levels, respectively than CC genotype (CC-hVSMC). EMSA assays showed that the polymorphism determines scarce amount of SRE-B/SREBP-2 complex formation and the failure of agLDL to further reduce these SRE-B/SREBP-2 complexes. Taken together, these results suggest that c.1-25C>G, by difficulting SREBP-2 binding, prevents SREBP-2 displacement required for LRP1 promoter response to LDL. In contrast, c.1-25C>G strongly favours Sp1/Sp3 binding and AngII-induced activity in Sp1/Sp3 dependent manner in GG-hVSMC. This increase is functionally translated into a higher capacity of GG-hVSMC to become foam cells from agLDL in presence of AngII. These results suggest that c.1-25C>G determines a lack of response to agLDL and an exacerbated response to AngII. It is thus conceivable that the presence of the polymorphism would be easily translated to vascular alterations in the presence of the pro-hypertensive autacoid, AngII.
Collapse
Affiliation(s)
- R Aledo
- Cardiovascular Research Center, CSIC-ICCC, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Autonomous University of Barcelona, Sant Antoni Mª Claret, 167, 08025 Barcelona, Spain
| | - P Costales
- Cardiovascular Research Center, CSIC-ICCC, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Autonomous University of Barcelona, Sant Antoni Mª Claret, 167, 08025 Barcelona, Spain
| | - C Ciudad
- Biochemistry and Molecular Biology Department, School of Pharmacy, IBUB, University of Barcelona, Barcelona, Spain
| | - V Noé
- Biochemistry and Molecular Biology Department, School of Pharmacy, IBUB, University of Barcelona, Barcelona, Spain
| | - V Llorente-Cortes
- Cardiovascular Research Center, CSIC-ICCC, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Autonomous University of Barcelona, Sant Antoni Mª Claret, 167, 08025 Barcelona, Spain.
| | - L Badimon
- Cardiovascular Research Center, CSIC-ICCC, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Autonomous University of Barcelona, Sant Antoni Mª Claret, 167, 08025 Barcelona, Spain
| |
Collapse
|
21
|
Guo G, Rödelsperger C, Digweed M, Robinson PN. Regulation of fibrillin-1 gene expression by Sp1. Gene 2013; 527:448-55. [PMID: 23860323 DOI: 10.1016/j.gene.2013.07.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2013] [Revised: 07/01/2013] [Accepted: 07/05/2013] [Indexed: 11/28/2022]
Abstract
Mutations in the fibrillin-1 gene (FBN1) cause Marfan Syndrome (MFS), a hereditary disorder of connective tissue. The transcription of FBN1 has been reported to be driven by a short ultraconserved region (SUPR) in the 5' untranslated exon A of FBN1, but the nature of other factors involved in FBN1 gene regulation has not been clarified. In this study, we characterized the transcription factors involved in FBN1 gene regulation. The results show that Sp1 protein binds to two putative binding sites in the promoter of FBN1. Overexpression of Sp1 resulted in a significant increase in both promoter activity and FBN1 mRNA level in HEK 293 cells, whereas inhibition or knockdown of Sp1 decreased FBN1 gene expression. In addition, we found that Poly [ADP-ribose] polymerase 1 (PARP1) binds to the palindromic sequence TCTCGCGAGA in the ultraconserved region of the FBN1 promoter and that the regulation of FBN1 expression by PARP1 is dependent on Sp1. These results indicate that both Sp1 and PARP1 contribute to FBN1 gene expression. These observations add to our understanding of the transcriptional regulation of FBN1 gene expression.
Collapse
Affiliation(s)
- Gao Guo
- Institute for Medical Genetics and Human Genetics, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | | | | | | |
Collapse
|
22
|
Xie L, Collins JF. Transcription factors Sp1 and Hif2α mediate induction of the copper-transporting ATPase (Atp7a) gene in intestinal epithelial cells during hypoxia. J Biol Chem 2013; 288:23943-52. [PMID: 23814049 DOI: 10.1074/jbc.m113.489500] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Genes with G/C-rich promoters were up-regulated in the duodenal epithelium of iron-deficient rats including those encoding iron (e.g. Dmt1 and Dcytb) and copper (e.g. Atp7a and Mt1) metabolism-related proteins. It was shown previously that an intestinal copper transporter (Atp7a) was co-regulated with iron transport-related genes by a hypoxia-inducible transcription factor, Hif2α. In the current study, we sought to test the role of Sp1 in transcriptional regulation of Atp7a expression during iron deprivation/hypoxia. Initial studies in IEC-6 cells showed that mithramycin, an Sp1 inhibitor, reduced expression of Atp7a and iron transport-related genes (Dmt1, Dcytb, and Fpn1) and blocked their induction by CoCl2, a hypoxia mimetic. Consistent with this, overexpression of Sp1 increased endogenous Atp7a mRNA and protein expression and stimulated Atp7a, Dmt1, and Dcytb promoter activity. Site-directed mutagenesis and functional analysis of a basal Atp7a promoter construct revealed four functional Sp1 binding sites that were necessary for Hif2α-mediated induction of promoter activity. Furthermore, chromatin immunoprecipitation (ChIP) assays confirmed that Sp1 specifically interacts with the Atp7a promoter in IEC-6 cells and in rat duodenal enterocytes. This investigation has thus revealed a novel aspect of Atp7a gene regulation in which Sp1 may be necessary for the HIF-mediated induction of gene transcription during iron deficiency/hypoxia. Understanding regulation of Atp7a expression may help further clarify the physiological role of copper in the maintenance of iron homeostasis. Furthermore, this Sp1/Hif2α regulatory mechanism may have broader implications for understanding the genetic response of the intestinal epithelium to maintain whole-body iron homeostasis during states of deficiency.
Collapse
Affiliation(s)
- Liwei Xie
- Food Science and Human Nutrition Department, University of Florida, Gainesville, Florida 32611, USA
| | | |
Collapse
|
23
|
Michinaga S, Ishida A, Takeuchi R, Koyama Y. Endothelin-1 stimulates cyclin D1 expression in rat cultured astrocytes via activation of Sp1. Neurochem Int 2013; 63:25-34. [PMID: 23619396 DOI: 10.1016/j.neuint.2013.04.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Revised: 03/25/2013] [Accepted: 04/08/2013] [Indexed: 11/16/2022]
Abstract
Endothelins (ETs), a family of vasoconstrictor peptides, are up-regulated in several pathological conditions in the brain, and induce astrocytic proliferation. We previously observed that ET-1 increased the expression of cyclin D1 protein. Thus, we confirmed the intracellular up-regulation of cyclin D1 by ET-1 in rat cultured astrocytes. Real-time PCR analysis indicated that ET-1 (100 nM) and Ala(1,3,11,15)-ET-1 (100 nM), a selective agonist of the ETB receptor, induced a time-dependent and transient increase in cyclin D1 mRNA. The effect of ET-1 was diminished by an ETB antagonist (1 μM BQ788) or inhibitors of Sp1 (500 nM mithramycin), ERK (50 μM PD98059), p38 (20 μM SB203580) and JNK (1 μM SP600125), but not inhibitors of NF-κB (10 μM SN50 and 100 μM pyrrolidine dithiocarbamate). The binding assay for Sp1 indicated that ET-1 increased the binding activity of Sp1 to consensus sequences, and two oligonucleotides of the cyclin D1 promoter including the Sp1-binding sites diminished the effect of ET-1. Western blot analysis showed that ET-1 induced time-dependent and transient phosphorylation of Sp1 on Thr453 and Thr739 via the ETB receptor. ET-1-induced phosphorylation of Sp1 was attenuated by PD98059 and SP600125. Additionally, ET-1 increased the incorporation of bromodeoxyuridine (BrdU) in cultured astrocytes and the number of BrdU-positive cells decreased in the presence of PD98059, SP600125 and mithramycin. These results suggest that ET-1 increases the expression of cyclin D1 via activation of Sp1 and induces astrocytic proliferation.
Collapse
Affiliation(s)
- Shotaro Michinaga
- Faculty of Pharmacy, Laboratory of Pharmacology, Osaka Ohtani University, 3-11-1 Nishikiori-Kita, Tonda-bayashi, Osaka 584-8540, Japan
| | | | | | | |
Collapse
|
24
|
Kajita Y, Kato T, Tamaki S, Furu M, Takahashi R, Nagayama S, Aoyama T, Nishiyama H, Nakamura E, Katagiri T, Nakamura Y, Ogawa O, Toguchida J. The transcription factor Sp3 regulates the expression of a metastasis-related marker of sarcoma, actin filament-associated protein 1-like 1 (AFAP1L1). PLoS One 2013; 8:e49709. [PMID: 23326307 PMCID: PMC3541374 DOI: 10.1371/journal.pone.0049709] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2012] [Accepted: 10/12/2012] [Indexed: 11/19/2022] Open
Abstract
We previously identified actin filament-associated protein 1-like 1 (AFAP1L1) as a metastasis-predicting marker from the gene-expression profiles of 65 spindle cell sarcomas, and demonstrated the up-regulation of AFAP1L1 expression to be an independent risk factor for distant metastasis in multivariate analyses. Little is known, however, about how the expression of AFAP1L1 is regulated. Luciferase reporter assays showed tandem binding motives of a specificity protein (Sp) located at −85 to −75 relative to the transcriptional start site to be essential to the promoter activity. Overexpression of Sp1 and Sp3 proteins transactivated the proximal AFAP1L1 promoter construct, and electrophoretic mobility shift assays showed that both Sp1 and Sp3 were able to bind to this region in vitro. Chromatin immunoprecipitation experiments, however, revealed that Sp3 is the major factor binding to the proximal promoter region of the AFAP1L1 gene in AFAP1L1- positive cells. Treatment with mithramycin A, an inhibitor of proteins binding to GC-rich regions, prevented Sp3 from binding to the proximal promoter region of AFAP1L1 and decreased its expression in a dose-dependent manner. Finally, knocking down Sp3 using small inhibitory RNA duplex (siRNA) reduced AFAP1L1 expression significantly, which was partially restored by expressing siRNA-resistant Sp3. These findings indicate a novel role for Sp3 in sarcomas as a driver for expression of the metastasis-related gene AFAP1L1.
Collapse
Affiliation(s)
- Yoichiro Kajita
- Department of Tissue Regeneration, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan
- Department of Urology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tomohisa Kato
- Department of Tissue Regeneration, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan
| | - Sakura Tamaki
- Department of Tissue Regeneration, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan
| | - Moritoshi Furu
- Department of Tissue Regeneration, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Ryo Takahashi
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Satoshi Nagayama
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tomoki Aoyama
- Department of Tissue Regeneration, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan
| | - Hiroyuki Nishiyama
- Department of Urology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Eijiro Nakamura
- Department of Urology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Toyomasa Katagiri
- Laboratory of Molecular Medicine, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yusuke Nakamura
- Laboratory of Molecular Medicine, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Osamu Ogawa
- Department of Urology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Junya Toguchida
- Department of Tissue Regeneration, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
- * E-mail:
| |
Collapse
|
25
|
Amodio N, Di Martino MT, Foresta U, Leone E, Lionetti M, Leotta M, Gullà AM, Pitari MR, Conforti F, Rossi M, Agosti V, Fulciniti M, Misso G, Morabito F, Ferrarini M, Neri A, Caraglia M, Munshi NC, Anderson KC, Tagliaferri P, Tassone P. miR-29b sensitizes multiple myeloma cells to bortezomib-induced apoptosis through the activation of a feedback loop with the transcription factor Sp1. Cell Death Dis 2012. [PMID: 23190608 PMCID: PMC3542610 DOI: 10.1038/cddis.2012.175] [Citation(s) in RCA: 126] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
MicroRNAs (miRNAs) with tumor-suppressor potential might have therapeutic applications in multiple myeloma (MM) through the modulation of still undiscovered molecular pathways. Here, we investigated the effects of enforced expression of miR-29b on the apoptotic occurrence in MM and highlighted its role in the context of a new transcriptional loop that is finely tuned by the proteasome inhibitor bortezomib. In details, in vitro growth inhibition and apoptosis of MM cells was induced by either transient expression of synthetic miR-29b or its stable lentivirus-enforced expression. We identified Sp1, a transcription factor endowed with oncogenic activity, as a negative regulator of miR-29b expression in MM cells. Since Sp1 expression and functions are regulated via the 26S proteasome, we investigated the effects of bortezomib on miR-29b-Sp1 loop, showing that miR-29b levels were indeed upregulated by the drug. At the same time, the bortezomib/miR-29b combination produced significant pro-apoptotic effects. We also demonstrated that the PI3K/AKT pathway plays a major role in the regulation of miR-29b-Sp1 loop and induction of apoptosis in MM cells. Finally, MM xenografts constitutively expressing miR-29b showed significant reduction of their tumorigenic potential. Our findings indicate that miR-29b is involved in a regulatory loop amenable of pharmacologic intervention and modulates the anti-MM activity of bortezomib in MM cells.
Collapse
Affiliation(s)
- N Amodio
- Medical Oncology, Department of Experimental and Clinical Medicine, Magna Graecia University and T Campanella Cancer Center, Salvatore Venuta Campus, Catanzaro, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Prospects and challenges for the development of new therapies for Ewing sarcoma. Pharmacol Ther 2012; 137:216-24. [PMID: 23085431 DOI: 10.1016/j.pharmthera.2012.10.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Accepted: 10/02/2012] [Indexed: 01/09/2023]
Abstract
The Ewing sarcoma family of tumors or Ewing sarcoma (ES) is the second most common malignant bone tumor of childhood. The prognosis for localized Ewing sarcoma has improved through the development of intense multimodal therapy over the past several decades. Unfortunately, patients with recurrent or metastatic disease continue to have a poor prognosis. Therefore, a number of complementary approaches are being developed in both the preclinical and clinical arenas to improve these outcomes. In this review, we will discuss efforts to directly target the biologic drivers of this disease and relate these efforts to the experience with several different agents both in the clinic and under development. We will review the data for compounds that have shown excellent activity in the clinic, such as the camptothecins, and summarize the biological data that supports this activity. In addition, we will review the clinical experience with IGF1 targeted agents, ET-743 and epigenetically targeted therapies, the substantial amount of literature that supports their activity in Ewing sarcoma and the challenges remaining translating these therapies to the clinic. Finally, we will highlight recent work aimed at directly targeting the EWS-FLI1 transcription factor with small molecules in Ewing tumors.
Collapse
|
27
|
Zip14 expression induced by lipopolysaccharides in macrophages attenuates inflammatory response. Inflamm Res 2012; 62:133-43. [DOI: 10.1007/s00011-012-0559-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Accepted: 09/18/2012] [Indexed: 11/30/2022] Open
|
28
|
Jiang Y, Fleet JC. Phorbol esters enhance 1α,25-dihydroxyvitamin D3-regulated 25-hydroxyvitamin D-24-hydroxylase (CYP24A1) gene expression through ERK-mediated phosphorylation of specific protein 3 (Sp3) in Caco-2 cells. Mol Cell Endocrinol 2012; 361:31-9. [PMID: 22871965 PMCID: PMC3414851 DOI: 10.1016/j.mce.2012.03.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2011] [Revised: 02/15/2012] [Accepted: 03/14/2012] [Indexed: 01/20/2023]
Abstract
Phorbol 12-myristate 13-acetate (PMA) increased 1,25(OH)(2)D(3)-induced human 25 hydroxyvitamin d-24 hydroxylase (hCYP24A1) gene expression and vitamin D receptor (VDR) binding to the hCYP24A1 promoter. It did not alter transient receptor potential cation channel, subfamily V, member 6 (TRPV6) expression, VDR binding to the TRPV6 promoter, or VDR binding to a crude chromatin preparation. PMA activated Extracellular signal-Regulated Kinases (ERK) 1/2 and p38 mitogen activated protein kinases (MAPK) and inhibiting these kinases reduced 1,25(OH)(2)D(3)-induced and PMA-enhanced hCYP24A1 promoter activity. Mithramycin A inhibits Specific Protein (Sp) family member binding to DNA and reduced 1,25(OH)(2)D(3)-induced and PMA-enhanced hCYP24A1 promoter activity. Sp1 or Sp3 siRNA knockdown reduced 1,25(OH)(2)D(3)-regulated hCYP24A1 promoter activity but only Sp3 siRNA reduced PMA-enhanced hCYP24A1 promoter activity. PMA increased MAPK-dependent Sp3 phosphorylation, Sp3-VDR interactions, and Sp3 binding to the hCYP24A1 promoter. These data suggest that MAPK signaling contributes to 1,25(OH)(2)D(3)-induced and PMA-enhanced CYP24A1 gene transcription by modulating Sp3 function.
Collapse
Affiliation(s)
- Yan Jiang
- Department of Nutrition Science, Purdue University, West Lafayette, IN 47907-2059, United States.
| | | |
Collapse
|
29
|
Sakuma K, Chen GY, Aoki M, Kannagi R. Induction of 6-sulfated glycans with cell adhesion activity via T-bet and GATA-3 in human helper T cells. BIOCHIMICA ET BIOPHYSICA ACTA 2012; 1820:841-8. [PMID: 22446378 DOI: 10.1016/j.bbagen.2012.03.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2011] [Revised: 03/07/2012] [Accepted: 03/08/2012] [Indexed: 02/08/2023]
Abstract
BACKGROUND Cell surface 6-sulfated glycans play important roles in various immunological events through cell-to-cell interactions. The 6-sulfation process is mediated by 6-sulfotransferase family isoenzymes. We previously demonstrated that GlcNAc6ST-1, one of the isoenzyme genes, is induced by GATA-3 and NF-κB in human helper T (Th) cells. However, transcriptional regulation of HEC-GlcNAc6ST, another isoenzyme important in Th cells, remains unclear. METHODS 5'-RACE analysis, chromatin immunoprecipitation, and reporter assays were performed to reveal transcriptional regulation of HEC-GlcNAc6ST. RNA-knockdown and forced expression experiments were performed to demonstrate the contribution of HEC-GlcNAc6ST to the 6-sulfated glycan expression. RESULTS We identified potential binding sites of Sp1, T-bet, and GATA-3 in the HEC-GlcNAc6ST promoter. Reporter assays indicated that transfection of Sp1 enhanced the activity, whereas mithramycin A, an Sp1-specific inhibitor, repressed it. Transfection of T-bet increased the activity, which was inhibited by introducing a mutation into the potential T-bet binding site. GATA-3 alone could not elevate the activity, although co-transfection of protein kinase A, which is known to enhance IL-5 transcription in Th2 cells through phosphorylation of GATA-3, caused elevation. RNA-knockdown and forced expression of HEC-GlcNAc6ST in Jurkat cells down- and up-regulated α2,6-sialylated 6-sulfo N-acetyllactosamine, a preferential ligand for B-cell-specific CD22 antigen, respectively. From these results, we concluded that T-bet and GATA-3 as well as Sp1 control the expression of glycan with cell-adhesion activity by regulating HEC-GlcNAc6ST transcription in Th cells. GENERAL SIGNIFICANCE These results may provide a clue to biological regulation of Th-cell interaction with selectins and other carbohydrate-recognition molecules by T-bet and GATA-3.
Collapse
Affiliation(s)
- Keiichiro Sakuma
- Division of Molecular Pathology, Aichi Cancer Center, Nagoya, Aichi 464-8681, Japan
| | | | | | | |
Collapse
|
30
|
Narumi K, Kobayashi M, Otake S, Furugen A, Takahashi N, Ogura J, Itagaki S, Hirano T, Yamaguchi H, Iseki K. Regulation of human monocarboxylate transporter 4 in skeletal muscle cells: The role of protein kinase C (PKC). Int J Pharm 2012; 428:25-32. [DOI: 10.1016/j.ijpharm.2012.02.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2011] [Revised: 01/30/2012] [Accepted: 02/10/2012] [Indexed: 11/16/2022]
|
31
|
c-Jun induction is independent of early growth response factor during cerebellar granule neuron apoptosis. Neuroreport 2012; 23:67-72. [DOI: 10.1097/wnr.0b013e32834e7d69] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
32
|
Transcription factor Sp1 is involved in expressional regulation of coxsackie and adenovirus receptor in cancer cells. J Biomed Biotechnol 2011; 2011:636497. [PMID: 22190856 PMCID: PMC3228682 DOI: 10.1155/2011/636497] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2011] [Revised: 09/17/2011] [Accepted: 10/07/2011] [Indexed: 12/14/2022] Open
Abstract
Coxsackie and adenovirus receptor (CAR) was first known as a virus receptor. Recently, it is also known to have tumor suppressive activity such as inhibition of cell proliferation, migration, and invasion. It is important to understand how CAR expression can be regulated in cancers. Based on an existence of putative Sp1 binding site within CAR promoter, we investigated whether indeed Sp1 is involved in the regulation of CAR expression. We observed that deletion or mutation of Sp1 binding motif (−503/−498) prominently impaired the Sp1 binding affinity and activity of CAR promoter. Histone deacetylase inhibitor (TSA) treatment enhanced recruitment of Sp1 to the CAR promoter in ChIP assay. Meanwhile, Sp1 binding inhibitor suppressed the recruitment. Exogenous expression of wild-type Sp1 increased CAR expression in CAR-negative cells; meanwhile, dominant negative Sp1 decreased the CAR expression in CAR-positive cells. These results indicate that Sp1 is involved in regulation of CAR expression.
Collapse
|
33
|
Rüegg J, Cai W, Karimi M, Kiss NB, Swedenborg E, Larsson C, Ekström TJ, Pongratz I. Epigenetic regulation of glucose transporter 4 by estrogen receptor β. Mol Endocrinol 2011; 25:2017-28. [PMID: 22016564 DOI: 10.1210/me.2011-1054] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Glucose transporter 4 (Glut4) is an important regulator of cellular glucose uptake in adipose tissue and skeletal muscle. The estrogen receptors α and β (ERα and ERβ) have been shown to regulate Glut4. However, the regulatory mechanisms are unclear, and there are conflicting results about the effects of the two ER isoforms on Glut4 activity. In this study we investigated how the lack of either ER isoform affects Glut4 expression in differentiated mouse embryonic fibroblasts. Our results demonstrate that Glut4 transcription is markedly reduced in cells lacking ERβ, both basally and upon induction by liver X receptor. These changes in Glut4 expression could not be explained by the lack of ERβ as ligand-activated transcription factor. They were rather brought about by hypermethylation of one single CpG in the Glut4 promoter in the ERβ-deficient cells. This CpG is part of an Sp1-binding site, and Sp1 binding was reduced by its methylation. Treatment with Sp1 inhibitor diminished Glut4 expression in wild-type, but not in ERβ-deficient cells, suggesting that reduced recruitment of Sp1 to the Glut4 promoter is responsible for the differences in Glut4 expression. Reintroduction of ERβ into ERβ-deficient cells partly restored Glut4 transcription and stabilized low DNA methylation after treatment with the DNA demethylating agent 5-Aza-2'-deoxycytidine. Our findings demonstrate the involvement of DNA methylation in Glut4 regulation and imply a novel function for ERβ in mediating epigenetic events and thereby regulating gene expression.
Collapse
Affiliation(s)
- Joëlle Rüegg
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, 141 57 Sweden.
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Sp1king out cancer (....and fibrosis?). J Cell Commun Signal 2011; 6:61-2. [PMID: 21822787 DOI: 10.1007/s12079-011-0147-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2011] [Accepted: 07/20/2011] [Indexed: 10/17/2022] Open
Abstract
It is becoming increasingly apparent that many of the basic mechanisms underlying cancers also underlie fibrotic diseases. For example, the Sp1 family of transcription factors plays an essential role in controlling the gene expression of proteins that promote both oncogenesis and fibrogenesis. The drug mithramycin, which prevents Sp1 binding to DNA, has been in use clinically for some cancers, but has side-effects. However, other drugs exist that affect Sp1 activity through promoting Sp1 protein degradation. Evidence has emerged that low levels of mithramycin can be combined with these drugs to result in potent antitumorigenic effects without resulting in obvious toxicity (Gao et al. Cancer Res 2011 Jun 20; Jia et al. Cancer Res 70:1111-1119, 2010). Given that Sp1 proteins also promote expression of profibrotic genes such as collagen type I and CCN2, it is possible that this combinatorial approach may be taken in the future to block not only cancer but also fibrosis.
Collapse
|
35
|
Transcriptional activation of TINF2, a gene encoding the telomere-associated protein TIN2, by Sp1 and NF-κB factors. PLoS One 2011; 6:e21333. [PMID: 21731707 PMCID: PMC3121743 DOI: 10.1371/journal.pone.0021333] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2011] [Accepted: 05/28/2011] [Indexed: 11/19/2022] Open
Abstract
The expression of the telomere-associated protein TIN2 has been shown to be essential for early embryonic development in mice and for development of a variety of human malignancies. Recently, germ-line mutations in TINF2, which encodes for the TIN2 protein, have been identified in a number of patients with bone-marrow failure syndromes. Yet, the molecular mechanisms that regulate TINF2 expression are largely unknown. To elucidate the mechanisms involved in human TINF2 regulation, we cloned a 2.7 kb genomic DNA fragment containing the putative promoter region and, through deletion analysis, identified a 406 bp region that functions as a minimal promoter. This promoter proximal region is predicted to contain several putative Sp1 and NF-κB binding sites based on bioinformatic analysis. Direct binding of the Sp1 and NF-κB transcription factors to the TIN2 promoter sequence was demonstrated by electrophoretic mobility shift assay (EMSA) and/or chromatin immunoprecipitation (ChIP) assays. Transfection of a plasmid carrying the Sp1 transcription factor into Sp-deficient SL2 cells strongly activated TIN2 promoter-driven luciferase reporter expression. Similarly, the NF-κB molecules p50 and p65 were found to strongly activate luciferase expression in NF-κB knockout MEFs. Mutating the predicted transcription factor binding sites effectively reduced TIN2 promoter activity. Various known chemical inhibitors of Sp1 and NF-κB could also strongly inhibit TIN2 transcriptional activity. Collectively, our results demonstrate the important roles that Sp1 and NF-κB play in regulating the expression of the human telomere-binding protein TIN2, which can shed important light on its possible role in causing various forms of human diseases and cancers.
Collapse
|
36
|
A protein-based electrochemical method for label-free characterization of sequence-specific protein–DNA interactions. Electrochim Acta 2011. [DOI: 10.1016/j.electacta.2011.04.050] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
37
|
Abstract
The proapoptotic BH3-only protein Bim is a crucial regulator of neuronal apoptosis. Previous studies have indicated the involvement of the c-Jun, FOXO1/3a, and B/C-Myb transcription factors in the regulation of Bim during neuronal apoptosis. However, the mechanism underlying the transcriptional regulation of Bim in activity deprivation-induced neuronal apoptosis has remained unclear. The present study demonstrates that early growth response 1 (Egr-1), rather than c-Jun, FOXO1/3a, or B/C-Myb, directly transactivates Bim gene expression to mediate apoptosis of rat cerebellar granule neurons. We showed that Egr-1 was sufficient and necessary for neuronal apoptosis. Suppression of Egr-1 activity using dominant-negative mutant or knockdown of Egr-1 using small interfering RNAs led to a decrease in Bim expression, whereas overexpression of Egr-1 resulted in induction of Bim. Deletion and site-directed mutagenesis of the Bim promoter revealed that Bim transcriptional activation depends primarily on a putative Egr-binding sequence between nucleotides -56 and -47 upstream of the start site. We also showed that Egr-1 binding to this sequence increased in response to activity deprivation in vitro and in vivo. Moreover, inhibition of Egr-1 binding to the Bim promoter, by mithramycin A and chromomycin A3, reduced the activity deprivation-induced increases in Bim promoter activity and mRNA and protein levels and protected neurons from apoptosis, further supporting the Egr-1-mediated transactivation of Bim. Additionally, Bim overcame the Egr-1 knockdown-mediated inhibition of apoptosis, whereas Bim knockdown impaired the increase in apoptosis induced by Egr-1. These findings establish Bim as an Egr-1 target gene in neurons, uncovering a novel Egr-1/Bim pathway by which activity deprivation induces neuronal apoptosis.
Collapse
|
38
|
Jin H, Kanthasamy A, Anantharam V, Rana A, Kanthasamy AG. Transcriptional regulation of pro-apoptotic protein kinase Cdelta: implications for oxidative stress-induced neuronal cell death. J Biol Chem 2011; 286:19840-59. [PMID: 21467032 DOI: 10.1074/jbc.m110.203687] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
We previously demonstrated that protein kinase Cδ (PKCδ; PKC delta) is an oxidative stress-sensitive kinase that plays a causal role in apoptotic cell death in neuronal cells. Although PKCδ activation has been extensively studied, relatively little is known about the molecular mechanisms controlling PKCδ expression. To characterize the regulation of PKCδ expression, we cloned an ∼2-kbp 5'-promoter segment of the mouse Prkcd gene. Deletion analysis indicated that the noncoding exon 1 region contained multiple Sp sites, including four GC boxes and one CACCC box, which directed the highest levels of transcription in neuronal cells. In addition, an upstream regulatory region containing adjacent repressive and anti-repressive elements with opposing regulatory activities was identified within the region -712 to -560. Detailed mutagenesis studies revealed that each Sp site made a positive contribution to PKCδ promoter expression. Overexpression of Sp family proteins markedly stimulated PKCδ promoter activity without any synergistic transactivating effect. Furthermore, experiments in Sp-deficient SL2 cells indicated long isoform Sp3 as the essential activator of PKCδ transcription. Importantly, both PKCδ promoter activity and endogenous PKCδ expression in NIE115 cells and primary striatal cultures were inhibited by mithramycin A. The results from chromatin immunoprecipitation and gel shift assays further confirmed the functional binding of Sp proteins to the PKCδ promoter. Additionally, we demonstrated that overexpression of p300 or CREB-binding protein increases the PKCδ promoter activity. This stimulatory effect requires intact Sp-binding sites and is independent of p300 histone acetyltransferase activity. Finally, modulation of Sp transcriptional activity or protein level profoundly altered the cell death induced by oxidative insult, demonstrating the functional significance of Sp-dependent PKCδ gene expression. Collectively, our findings may have implications for development of new translational strategies against oxidative damage.
Collapse
Affiliation(s)
- Huajun Jin
- Parkinson's Disorder Research Laboratory, Iowa Center for Advanced Neurotoxicology, Department of Biomedical Sciences, Iowa State University, Ames, Iowa 50011, USA
| | | | | | | | | |
Collapse
|
39
|
Beaulieu E, Green L, Elsby L, Alourfi Z, Morand EF, Ray DW, Donn R. Identification of a novel cell type-specific intronic enhancer of macrophage migration inhibitory factor (MIF) and its regulation by mithramycin. Clin Exp Immunol 2010; 163:178-88. [PMID: 21087445 PMCID: PMC3043308 DOI: 10.1111/j.1365-2249.2010.04289.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The aim of this study was to determine the genetic regulation of macrophage migration inhibitory factor (MIF). DNase I hypersensitivity was used to identify potential hypersensitive sites (HS) across the MIF gene locus. Reporter gene assays were performed in different human cell lines with constructs containing the native or mutated HS element. Following phylogenetic and transcription factor binding profiling, electrophoretic mobility shift assay (EMSA) and RNA interference were performed and the effects of incubation with mithramycin, an antibiotic that binds GC boxes, were also studied. An HS centred on the first intron of MIF was identified. The HS acted as an enhancer in human T lymphoblasts (CEMC7A), human embryonic kidney cells (HEK293T) and human monocytic cells (THP-1), but not in a fibroblast-like synoviocyte (FLS) cell line (SW982) or cultured FLS derived from rheumatoid arthritis (RA) patients. Two cis-elements within the first intron were found to be responsible for the enhancer activity. Mutation of the consensus Sp1 GC box on each cis-element abrogated enhancer activity and EMSA indicated Sp1 binding to one of the cis-elements contained in the intron. SiRNA knock-down of Sp1 alone or Sp1 and Sp3 together was incomplete and did not alter the enhancer activity. Mithramycin inhibited expression of MIF in CEMC7A cells. This effect was specific to the intronic enhancer and was not seen on the MIF promoter. These results identify a novel, cell type-specific enhancer of MIF. The enhancer appears to be driven by Sp1 or related Sp family members and is highly sensitive to inhibition via mithramycin.
Collapse
|
40
|
García B, González-Sabín J, Menéndez N, Braña AF, Núñez LE, Morís F, Salas JA, Méndez C. The chromomycin CmmA acetyltransferase: a membrane-bound enzyme as a tool for increasing structural diversity of the antitumour mithramycin. Microb Biotechnol 2010; 4:226-38. [PMID: 21342468 PMCID: PMC3818863 DOI: 10.1111/j.1751-7915.2010.00229.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Mithramycin and chromomycin A3 are two structurally related antitumour compounds, which differ in the glycosylation profiles and functional group substitutions of the sugars. Chromomycin contains two acetyl groups, which are incorporated during the biosynthesis by the acetyltransferase CmmA in Streptomyces griseus ssp. griseus. A bioconversion strategy using an engineered S. griseus strain generated seven novel acetylated mithramycins. The newly formed compounds were purified and characterized by MS and NMR. These new compounds differ from their parental compounds in the presence of one, two or three acetyl groups, attached at 3E, 4E and/or 4D positions. All new mithramycin analogues showed antitumour activity at micromolar of lower concentrations. Some of the compounds showed improved activities against glioblastoma or pancreas tumour cells. The CmmA acetyltransferase was located in the cell membrane and was shown to accept several acyl‐CoA substrates. All these results highlight the potential of CmmA as a tool to create structural diversity in these antitumour compounds.
Collapse
Affiliation(s)
- Beatriz García
- Departamento de Biología Funcional e Instituto Universitario de Oncología del Principado de Asturias (I.U.O.P.A), Universidad de Oviedo, Oviedo, Spain
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Transcriptional control of KCNQ channel genes and the regulation of neuronal excitability. J Neurosci 2010; 30:13235-45. [PMID: 20926649 DOI: 10.1523/jneurosci.1981-10.2010] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Regulation of the resting membrane potential and the repolarization of neurons are important in regulating neuronal excitability. The potassium channel subunits Kv7.2 and Kv7.3 play a key role in stabilizing neuronal activity. Mutations in KCNQ2 and KCNQ3, the genes encoding Kv7.2 and Kv7.3, cause a neonatal form of epilepsy, and activators of these channels have been identified as novel antiepileptics and analgesics. Despite the observations that regulation of these subunits has profound effects on neuronal function, almost nothing is known about the mechanisms responsible for controlling appropriate expression levels. Here we identify two mechanisms responsible for regulating KCNQ2 and KCNQ3 mRNA levels. We show that the transcription factor Sp1 activates expression of both KCNQ2 and KCNQ3, whereas the transcriptional repressor REST (repressor element 1-silencing transcription factor) represses expression of both of these genes. Furthermore, we show that transcriptional regulation of KCNQ genes is mirrored by the correlated changes in M-current density and excitability of native sensory neurons. We propose that these mechanisms are important in the control of excitability of neurons and may have implications in seizure activity and pain.
Collapse
|
42
|
Iancu OD, Darakjian P, Walter NAR, Malmanger B, Oberbeck D, Belknap J, McWeeney S, Hitzemann R. Genetic diversity and striatal gene networks: focus on the heterogeneous stock-collaborative cross (HS-CC) mouse. BMC Genomics 2010; 11:585. [PMID: 20959017 PMCID: PMC3091732 DOI: 10.1186/1471-2164-11-585] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2010] [Accepted: 10/19/2010] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND The current study focused on the extent genetic diversity within a species (Mus musculus) affects gene co-expression network structure. To examine this issue, we have created a new mouse resource, a heterogeneous stock (HS) formed from the same eight inbred strains that have been used to create the collaborative cross (CC). The eight inbred strains capture > 90% of the genetic diversity available within the species. For contrast with the HS-CC, a C57BL/6J (B6) × DBA/2J (D2) F2 intercross and the HS4, derived from crossing the B6, D2, BALB/cJ and LP/J strains, were used. Brain (striatum) gene expression data were obtained using the Illumina Mouse WG 6.1 array, and the data sets were interrogated using a weighted gene co-expression network analysis (WGCNA). RESULTS Genes reliably detected as expressed were similar in all three data sets as was the variability of expression. As measured by the WGCNA, the modular structure of the transcriptome networks was also preserved both on the basis of module assignment and from the perspective of the topological overlap maps. Details of the HS-CC gene modules are provided; essentially identical results were obtained for the HS4 and F2 modules. Gene ontology annotation of the modules revealed a significant overrepresentation in some modules for neuronal processes, e.g., central nervous system development. Integration with known protein-protein interactions data indicated significant enrichment among co-expressed genes. We also noted significant overlap with markers of central nervous system cell types (neurons, oligodendrocytes and astrocytes). Using the Allen Brain Atlas, we found evidence of spatial co-localization within the striatum for several modules. Finally, for some modules it was possible to detect an enrichment of transcription binding sites. The binding site for Wt1, which is associated with neurodegeneration, was the most significantly overrepresented. CONCLUSIONS Despite the marked differences in genetic diversity, the transcriptome structure was remarkably similar for the F2, HS4 and HS-CC. These data suggest that it should be possible to integrate network data from simple and complex crosses. A careful examination of the HS-CC transcriptome revealed the expected structure for striatal gene expression. Importantly, we demonstrate the integration of anatomical and network expression data.
Collapse
Affiliation(s)
- Ovidiu D Iancu
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA.
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Infection with E1B-mutant adenovirus stabilizes p53 but blocks p53 acetylation and activity through E1A. Oncogene 2010; 30:865-75. [PMID: 20935676 DOI: 10.1038/onc.2010.461] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Wild-type adenovirus type 5 eliminates p53 through the E1B-55kDa and E4-34kDa gene products. Deletion or mutation of E1B-55kDa has long been thought to confer p53-selective replication of oncolytic viruses. We show here that infection with E1B-defective adenovirus mutants induces massive accumulation of p53, without obvious defects in p53 localization, phosphorylation, conformation and oligomerization. Nonetheless, p53 completely failed to induce its target genes in this scenario, for example, p21/CDKN1A, Mdm2 and PUMA. Two regions of the E1A gene products independently contributed to the suppression of p21 transcription. Depending on the E1A conserved region 3, E1B-defective adenovirus impaired the ability of the transcription factor Sp1 to bind the p21 promoter. Moreover, the amino terminal region of E1A, binding the acetyl transferases p300 and CREB-binding protein, blocked p53 K382 acetylation in infected cells. Mutating either of these E1A regions, in addition to E1B, partially restored p21 mRNA levels. Our findings argue that adenovirus attenuates p53-mediated p21 induction, through at least two E1B-independent mechanisms. Other virus species and cancer cells may employ analogous strategies to impair p53 activity.
Collapse
|
44
|
Yu HT, Chan WWL, Chai KH, Lee CWC, Chang RCC, Yu MS, McLoughlin DM, Miller CCJ, Lau KF. Transcriptional regulation of human FE65, a ligand of Alzheimer's disease amyloid precursor protein, by Sp1. J Cell Biochem 2010; 109:782-93. [PMID: 20091743 DOI: 10.1002/jcb.22457] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
FE65 is a neuronal-enriched adaptor protein that binds to the Alzheimer's disease amyloid precursor protein (APP). FE65 forms a transcriptionally active complex with the APP intracellular domain (AICD). The precise gene targets for this complex are unclear but several Alzheimer's disease-linked genes have been proposed. Additionally, evidence suggests that FE65 influences APP metabolism. The mechanism by which FE65 expression is regulated is as yet unknown. To gain insight into the regulatory mechanism, we cloned a 1.6 kb fragment upstream of the human FE65 gene and found that it possesses particularly strong promoter activity in neurones. To delineate essential regions in the human FE65 promoter, a series of deletion mutants were generated. The minimal FE65 promoter was located between -100 and +5, which contains a functional Sp1 site. Overexpression of the transcription factor Sp1 potentiates the FE65 promoter activity. Conversely, suppression of the FE65 promoter was observed in cells either treated with an Sp1 inhibitor or in which Sp1 was knocked down. Furthermore, reduced levels of Sp1 resulted in downregulation of endogenous FE65 mRNA and protein. These findings reveal that Sp1 plays a crucial role in transcriptional control of the human FE65 gene.
Collapse
Affiliation(s)
- Hoi-Tin Yu
- Department of Biochemistry (Science), The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Liu S, Wu LC, Pang J, Santhanam R, Schwind S, Wu YZ, Hickey C, Yu J, Becker H, Maharry K, Radmacher MD, Li C, Whitman SP, Mishra A, Stauffer N, Eiring AM, Briesewitz R, Baiocchi RA, Chan KK, Paschka P, Caligiuri MA, Byrd JC, Croce CM, Bloomfield CD, Perrotti D, Garzon R, Marcucci G. Sp1/NFkappaB/HDAC/miR-29b regulatory network in KIT-driven myeloid leukemia. Cancer Cell 2010; 17:333-47. [PMID: 20385359 PMCID: PMC2917066 DOI: 10.1016/j.ccr.2010.03.008] [Citation(s) in RCA: 218] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2009] [Revised: 11/29/2009] [Accepted: 03/17/2010] [Indexed: 12/29/2022]
Abstract
The biologic and clinical significance of KIT overexpression that associates with KIT gain-of-function mutations occurring in subsets of acute myeloid leukemia (AML) (i.e., core binding factor AML) is unknown. Here, we show that KIT mutations lead to MYC-dependent miR-29b repression and increased levels of the miR-29b target Sp1 in KIT-driven leukemia. Sp1 enhances its own expression by participating in a NFkappaB/HDAC complex that further represses miR-29b transcription. Upregulated Sp1 then binds NFkappaB and transactivates KIT. Therefore, activated KIT ultimately induces its own transcription. Our results provide evidence that the mechanisms of Sp1/NFkappaB/HDAC/miR-29b-dependent KIT overexpression contribute to leukemia growth and can be successfully targeted by pharmacological disruption of the Sp1/NFkappaB/HDAC complex or synthetic miR-29b treatment in KIT-driven AML.
Collapse
Affiliation(s)
- Shujun Liu
- Divisions of Hematology-Oncology, The Ohio State University, Columbus, OH 4321
- The Comprehensive Cancer Center, The Ohio State University, Columbus, OH 4321
- To whom correspondence should be addressed: Shujun Liu and Guido Marcucci, The Ohio State University, 898 Biomedical Research Tower, 460 West 12th Avenue, Columbus, Ohio 43210. Phone: 614-293-7597. FAX: 614-293-7527. or
| | - Lai-Chu Wu
- Department of Molecular & Cellular Biochemistry, The Ohio State University, Columbus, OH 4321
| | - Jiuxia Pang
- The Comprehensive Cancer Center, The Ohio State University, Columbus, OH 4321
| | - Ramasamy Santhanam
- Divisions of Hematology-Oncology, The Ohio State University, Columbus, OH 4321
- The Comprehensive Cancer Center, The Ohio State University, Columbus, OH 4321
| | - Sebastian Schwind
- Divisions of Hematology-Oncology, The Ohio State University, Columbus, OH 4321
- The Comprehensive Cancer Center, The Ohio State University, Columbus, OH 4321
| | - Yue-Zhong Wu
- Divisions of Hematology-Oncology, The Ohio State University, Columbus, OH 4321
| | - Christopher Hickey
- Divisions of Hematology-Oncology, The Ohio State University, Columbus, OH 4321
| | - Jianhua Yu
- The Comprehensive Cancer Center, The Ohio State University, Columbus, OH 4321
- Department of Molecular Virology, Immunology and Cancer Genetics, The Ohio State University, Columbus, OH 4321
| | - Heiko Becker
- Divisions of Hematology-Oncology, The Ohio State University, Columbus, OH 4321
- The Comprehensive Cancer Center, The Ohio State University, Columbus, OH 4321
| | - Kati Maharry
- Divisions of Hematology-Oncology, The Ohio State University, Columbus, OH 4321
- The Comprehensive Cancer Center, The Ohio State University, Columbus, OH 4321
| | - Michael D Radmacher
- The Comprehensive Cancer Center, The Ohio State University, Columbus, OH 4321
| | - Chenglong Li
- Department of Molecular & Cellular Biochemistry, The Ohio State University, Columbus, OH 4321
| | - Susan P. Whitman
- The Comprehensive Cancer Center, The Ohio State University, Columbus, OH 4321
- Department of Molecular Virology, Immunology and Cancer Genetics, The Ohio State University, Columbus, OH 4321
| | - Anjali Mishra
- The Comprehensive Cancer Center, The Ohio State University, Columbus, OH 4321
- Department of Molecular Virology, Immunology and Cancer Genetics, The Ohio State University, Columbus, OH 4321
| | - Nicole Stauffer
- Divisions of Hematology-Oncology, The Ohio State University, Columbus, OH 4321
- The Comprehensive Cancer Center, The Ohio State University, Columbus, OH 4321
| | - Anna M. Eiring
- Department of Molecular Virology, Immunology and Cancer Genetics, The Ohio State University, Columbus, OH 4321
| | - Roger Briesewitz
- The Comprehensive Cancer Center, The Ohio State University, Columbus, OH 4321
| | - Robert A. Baiocchi
- Divisions of Hematology-Oncology, The Ohio State University, Columbus, OH 4321
- The Comprehensive Cancer Center, The Ohio State University, Columbus, OH 4321
| | - Kenneth K. Chan
- The Comprehensive Cancer Center, The Ohio State University, Columbus, OH 4321
- Division of Pharmaceutics of College of Pharmacy, The Ohio State University, Columbus, OH 4321
| | - Peter Paschka
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany
| | - Michael A. Caligiuri
- Divisions of Hematology-Oncology, The Ohio State University, Columbus, OH 4321
- The Comprehensive Cancer Center, The Ohio State University, Columbus, OH 4321
- Department of Molecular Virology, Immunology and Cancer Genetics, The Ohio State University, Columbus, OH 4321
| | - John C. Byrd
- Divisions of Hematology-Oncology, The Ohio State University, Columbus, OH 4321
- The Comprehensive Cancer Center, The Ohio State University, Columbus, OH 4321
- Department of Molecular Virology, Immunology and Cancer Genetics, The Ohio State University, Columbus, OH 4321
| | - Carlo M Croce
- The Comprehensive Cancer Center, The Ohio State University, Columbus, OH 4321
- Department of Molecular Virology, Immunology and Cancer Genetics, The Ohio State University, Columbus, OH 4321
| | - Clara D. Bloomfield
- Divisions of Hematology-Oncology, The Ohio State University, Columbus, OH 4321
- The Comprehensive Cancer Center, The Ohio State University, Columbus, OH 4321
| | - Danilo Perrotti
- The Comprehensive Cancer Center, The Ohio State University, Columbus, OH 4321
- Department of Molecular Virology, Immunology and Cancer Genetics, The Ohio State University, Columbus, OH 4321
| | - Ramiro Garzon
- Divisions of Hematology-Oncology, The Ohio State University, Columbus, OH 4321
- The Comprehensive Cancer Center, The Ohio State University, Columbus, OH 4321
| | - Guido Marcucci
- Divisions of Hematology-Oncology, The Ohio State University, Columbus, OH 4321
- The Comprehensive Cancer Center, The Ohio State University, Columbus, OH 4321
- Department of Molecular Virology, Immunology and Cancer Genetics, The Ohio State University, Columbus, OH 4321
- Division of Pharmaceutics of College of Pharmacy, The Ohio State University, Columbus, OH 4321
- To whom correspondence should be addressed: Shujun Liu and Guido Marcucci, The Ohio State University, 898 Biomedical Research Tower, 460 West 12th Avenue, Columbus, Ohio 43210. Phone: 614-293-7597. FAX: 614-293-7527. or
| |
Collapse
|
46
|
Vascular endothelial growth factor stimulates protein kinase CbetaII expression in chronic lymphocytic leukemia cells. Blood 2010; 115:4447-54. [PMID: 20164467 DOI: 10.1182/blood-2009-06-229872] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Chronic lymphocytic leukemia (CLL) is a malignant disease of mature B lymphocytes. We have previously shown that a characteristic feature of CLL cells are high levels of expression and activity of protein kinase CbetaII (PKCbetaII), and that this might influence disease progression by modulating signaling in response to B-cell receptor engagement. The aim of the present work was to investigate the factors involved in stimulating PKCbetaII expression in CLL cells. Here we show that the activation of PKCbetaII in CLL cells stimulated with vascular endothelial growth factor (VEGF) can drive expression of the gene for PKCbeta, PRKCB1. We found that this effect of VEGF on PRKCB1 transcription is paralleled by high expression of PKCbetaII protein and therefore probably contributes to the malignant phenotype of CLL cells. Taken together, the data presented in this study demonstrate that VEGF, in addition to its role in providing prosurvival signals, also plays a role in overexpression of PKCbetaII, an enzyme with a specific pathophysiologic role in CLL.
Collapse
|
47
|
Wilson AJ, Chueh AC, Tögel L, Corner GA, Ahmed N, Goel S, Byun DS, Nasser S, Houston MA, Jhawer M, Smartt HJM, Murray LB, Nicholas C, Heerdt BG, Arango D, Augenlicht LH, Mariadason JM. Apoptotic sensitivity of colon cancer cells to histone deacetylase inhibitors is mediated by an Sp1/Sp3-activated transcriptional program involving immediate-early gene induction. Cancer Res 2010; 70:609-20. [PMID: 20068171 DOI: 10.1158/0008-5472.can-09-2327] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Histone deacetylase inhibitors (HDACi) induce growth arrest and apoptosis in colon cancer cells and are being considered for colon cancer therapy. The underlying mechanism of action of these effects is poorly defined with both transcription-dependent and -independent mechanisms implicated. We screened a panel of 30 colon cancer cell lines for sensitivity to HDACi-induced apoptosis and correlated the differences with gene expression patterns induced by HDACi in the five most sensitive and resistant lines. A robust and reproducible transcriptional response involving coordinate induction of multiple immediate-early (fos, jun, egr1, egr3, atf3, arc, nr4a1) and stress response genes (Ndrg4, Mt1B, Mt1E, Mt1F, Mt1H) was selectively induced in HDACi sensitive cells. Notably, a significant percentage of these genes were basally repressed in colon tumors. Bioinformatics analysis revealed that the promoter regions of the HDACi-induced genes were enriched for KLF4/Sp1/Sp3 transcription factor binding sites. Altering KLF4 levels failed to modulate apoptosis or transcriptional responses to HDACi treatment. In contrast, HDACi preferentially stimulated the activity of Spl/Sp3 and blocking their action attenuated both the transcriptional and apoptotic responses to HDACi treatment. Our findings link HDACi-induced apoptosis to activation of a Spl/Sp3-mediated response that involves derepression of a transcriptional network basally repressed in colon cancer.
Collapse
Affiliation(s)
- Andrew J Wilson
- Department of Oncology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Mithramycin protects against dopaminergic neurotoxicity in the mouse brain after administration of methamphetamine. Brain Res 2009; 1301:189-96. [PMID: 19748494 DOI: 10.1016/j.brainres.2009.09.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2009] [Revised: 08/29/2009] [Accepted: 09/04/2009] [Indexed: 11/22/2022]
Abstract
The present study was undertaken to examine the effects of mithramycin, an inhibitor of transcription factor Specificity protein (Sp)-1, on the behavioral changes and dopaminergic neurotoxicity in the mouse striatum after administration of methamphetamine (METH). Pretreatment with mithramycin (75, 150 or 300 microg/kg) did not alter acute hyperlocomotion in mice after a single administration of METH (3 mg/kg). However, the development of behavioral sensitization in mice after repeated administration of METH (3 mg/kg/day, once daily for 5 days) was significantly blocked by pretreatment with mithramycin (300 microg/kg). Furthermore, pretreatment with mithramycin (300 microg/kg) significantly attenuated the hyperthermia in mice after repeated administration of METH (3 mg/kgx3, 3-h intervals). Moreover, the combination of pretreatment and subsequent administration of mithramycin (75, 150 or 300 microg/kg) significantly attenuated the reductions of dopamine (DA), its major metabolite 3,4-dihydroxyphenylacetic acid (DOPAC) and DA transporter (DAT) in the striatum after repeated administration of METH (3 mg/kgx3, 3-h intervals), and these attenuations were dose dependent. These findings suggest that mithramycin attenuates the development of behavioral sensitization and dopaminergic neurotoxicity in mice after repeated administration of METH. Therefore, mithramycin could have potential for the treatment of METH abusers, particularly since this drug has been approved by the Food and Drug Administration in the United States. In the future, however, another Sp1 inhibitors with fewer side effects might be more appropriate.
Collapse
|
49
|
Okuda T, Furukawa K, Nakayama KI. A novel, promoter-based, target-specific assay identifies 2-deoxy-D-glucose as an inhibitor of globotriaosylceramide biosynthesis. FEBS J 2009; 276:5191-202. [PMID: 19674101 DOI: 10.1111/j.1742-4658.2009.07215.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Abnormal biosynthesis of globotriaosylceramide (Gb3) is known to be associated with Gb3-related diseases, such as Fabry disease. The Gb3 synthase gene (Gb3S) codes for alpha1,4-galactosyltransferase, which is a key enzyme involved in Gb3 biosynthesis in vivo. Transcriptional repression of Gb3S is a way to control Gb3 biosynthesis and may be a suitable target for the treatment of Gb3-related diseases. To find a transcriptional inhibitor for Gb3S, we developed a convenient cell-based chemical screening assay system by constructing a fusion gene construct of the human Gb3S promoter and a secreted luciferase as reporter. Using this assay, we identified 2-deoxy-D-glucose as a potent inhibitor for the Gb3S promoter. In cultured cells, 2-deoxy-D-glucose markedly reduced endogenous Gb3S mRNA levels, resulting in a reduction in cellular Gb3 content and a corresponding accumulation of the precursor lactosylceramide. Moreover, cytokine-induced expression of Gb3 on the cell surface of endothelial cells, which is closely related to the onset of hemolytic uremic syndrome in O157-infected patients, was also suppressed by 2-deoxy-D-glucose treatment. These results indicate that 2-deoxy-D-glucose can control Gb3 biosynthesis through the inhibition of Gb3S transcription. Furthermore, we demonstrated the general utility of our novel screening assay for the identification of new inhibitors of glycosphingolipid biosynthesis.
Collapse
Affiliation(s)
- Tetsuya Okuda
- Glycolipids Function Analysis Team, Health Technology Research Center, National Institute of Advanced Industrial Science and Technology, Kagawa, Japan.
| | | | | |
Collapse
|
50
|
Sun X, Ritzenthaler JD, Zhong X, Zheng Y, Roman J, Han S. Nicotine stimulates PPARbeta/delta expression in human lung carcinoma cells through activation of PI3K/mTOR and suppression of AP-2alpha. Cancer Res 2009; 69:6445-53. [PMID: 19654299 DOI: 10.1158/0008-5472.can-09-1001] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
We previously showed that nicotine stimulates non-small cell lung carcinoma (NSCLC) cell proliferation through nicotinic acetylcholine receptor (nAChR)-mediated signals. Activation of peroxisome proliferator-activated receptor beta/delta (PPARbeta/delta) has also been shown to induce NSCLC cell growth. Here, we explore the potential link between nicotine and PPARbeta/delta and report that nicotine increases the expression of PPARbeta/delta protein; this effect was blocked by an alpha7 nAChR antagonist (alpha-bungarotoxin), by alpha7 nAChR short interfering RNA, and by inhibitors of phosphatidylinositol 3-kinase (PI3K; wortmannin and LY294002) and mammalian target of rapamycin (mTOR; rapamycin). In contrast, this effect was enhanced by PUN282987, an alpha7 nAChR agonist. Silencing of PPARbeta/delta attenuated the stimulatory effect of nicotine on cell growth, which was overcome by transfection of an exogenous PPARbeta/delta expression vector. Of note, nicotine induced complex formation between alpha7 nAChR and PPARbeta/delta protein and increased PPARbeta/delta gene promoter activity through inhibition of AP-2alpha as shown by reduced AP-2alpha binding using electrophoretic gel mobility shift and chromatin immunoprecipitation assays. In addition, silencing of Sp1 attenuated the effect of nicotine on PPARbeta/delta. Collectively, our results show that nicotine increases PPARbeta/delta gene expression through alpha7 nAChR-mediated activation of PI3K/mTOR signals that inhibit AP-2alpha protein expression and DNA binding activity to the PPARbeta/delta gene promoter. Sp1 seems to modulate this process. This study unveils a novel mechanism by which nicotine promotes human lung carcinoma cell growth.
Collapse
Affiliation(s)
- XiaoJuan Sun
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | | | | | | | | |
Collapse
|