1
|
Dulce RA, Hatzistergos KE, Kanashiro-Takeuchi RM, Takeuchi LM, Balkan W, Hare JM. Growth hormone-releasing hormone signaling and manifestations within the cardiovascular system. Rev Endocr Metab Disord 2025:10.1007/s11154-024-09939-0. [PMID: 39883351 DOI: 10.1007/s11154-024-09939-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/23/2024] [Indexed: 01/31/2025]
Abstract
Growth hormone (GH)-releasing hormone (GHRH), a hypothalamic peptide initially characterized for its role in GH regulation, has gained increasing attention due to its GH-independent action on peripheral physiology, including that of the cardiovascular system. While its effects on the peripheral vasculature are still under investigation, GHRH and synthetic agonists have exhibited remarkable receptor-mediated cardioprotective properties in preclinical models. GHRH and its analogs enhance myocardial function by improving contractility, reducing oxidative stress, inflammation, and offsetting pathological remodeling. Studies performed in small and large animal models have demonstrated the efficacy of these compounds in diverse cardiomyopathies, suggesting their potential as promising therapeutic agents. However, the clinical translation of GHRH synthetic analogs still faces challenges related to the route of administration and potential side effects mainly associated with activation of the GH/IGF-I axis. Despite these hurdles, the compelling evidence supporting their role in cardiac repair makes GHRH analogs attractive candidates for clinical testing in the treatment of various cardiac diseases.
Collapse
Affiliation(s)
- Raul A Dulce
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Biomedical Research Building, 1501 N.W. 10th Avenue, Room 908, Miami, FL, 33136, USA
| | - Konstantinos E Hatzistergos
- Department of Genetics, Development and Molecular Biology, School of Biology, Aristotle University of Thessaloniki, Thessaloniki, 54124, Greece
| | - Rosemeire M Kanashiro-Takeuchi
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Biomedical Research Building, 1501 N.W. 10th Avenue, Room 908, Miami, FL, 33136, USA
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Lauro M Takeuchi
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Biomedical Research Building, 1501 N.W. 10th Avenue, Room 908, Miami, FL, 33136, USA
| | - Wayne Balkan
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Biomedical Research Building, 1501 N.W. 10th Avenue, Room 908, Miami, FL, 33136, USA
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Joshua M Hare
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Biomedical Research Building, 1501 N.W. 10th Avenue, Room 908, Miami, FL, 33136, USA.
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
| |
Collapse
|
2
|
Mzizi Y, Mbambara S, Moetlhoa B, Mahapane J, Mdanda S, Sathekge M, Kgatle M. Ionising radiation exposure-induced regulation of selected biomarkers and their impact in cancer and treatment. FRONTIERS IN NUCLEAR MEDICINE (LAUSANNE, SWITZERLAND) 2024; 4:1469897. [PMID: 39498386 PMCID: PMC11532091 DOI: 10.3389/fnume.2024.1469897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 09/30/2024] [Indexed: 11/07/2024]
Abstract
Ionising radiation (IR) is a form of energy that travels as electromagnetic waves or particles. While it is vital in medical and occupational health settings, IR can also damage DNA, leading to mutations, chromosomal aberrations, and transcriptional changes that disrupt the functions of certain cell regulators, genes, and transcription factors. These disruptions can alter functions critical for cancer development, progression, and treatment response. Additionally, IR can affect various cellular proteins and their regulators within different cell signalling pathways, resulting in physiological changes that may promote cancer development, progression, and resistance to treatment. Understanding these impacts is crucial for developing strategies to mitigate the harmful effects of IR exposure and improve cancer treatment outcomes. This review focuses on specific genes and protein biomarkers regulated in response to chronic IR exposure, and how their regulation impacts disease onset, progression, and treatment response.
Collapse
Affiliation(s)
- Yonwaba Mzizi
- Department of Nuclear Medicine, University of Pretoria and Steve Biko Academic Hospital, Pretoria, South Africa
- Basic and Translational Research, Nuclear Medicine Research Infrastructure (NuMeRI), Steve Biko Academic Hospital, Pretoria, South Africa
| | - Saidon Mbambara
- Department of Nuclear Medicine, University of Pretoria and Steve Biko Academic Hospital, Pretoria, South Africa
- Basic and Translational Research, Nuclear Medicine Research Infrastructure (NuMeRI), Steve Biko Academic Hospital, Pretoria, South Africa
- Department of Biomedical Sciences, Tropical Diseases Research Centre, Ndola, Zambia
| | - Boitumelo Moetlhoa
- Basic and Translational Research, Nuclear Medicine Research Infrastructure (NuMeRI), Steve Biko Academic Hospital, Pretoria, South Africa
- School of Health Systems and Public Health, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Johncy Mahapane
- Department of Radiography, University of Pretoria, Pretoria, South Africa
| | - Sipho Mdanda
- Department of Nuclear Medicine, University of Pretoria and Steve Biko Academic Hospital, Pretoria, South Africa
- Basic and Translational Research, Nuclear Medicine Research Infrastructure (NuMeRI), Steve Biko Academic Hospital, Pretoria, South Africa
| | - Mike Sathekge
- Department of Nuclear Medicine, University of Pretoria and Steve Biko Academic Hospital, Pretoria, South Africa
- Basic and Translational Research, Nuclear Medicine Research Infrastructure (NuMeRI), Steve Biko Academic Hospital, Pretoria, South Africa
| | - Mankgopo Kgatle
- Department of Nuclear Medicine, University of Pretoria and Steve Biko Academic Hospital, Pretoria, South Africa
- Basic and Translational Research, Nuclear Medicine Research Infrastructure (NuMeRI), Steve Biko Academic Hospital, Pretoria, South Africa
- Department of Medicine, University of Cape Town and Groote Schuur Hospital, Cape Town, South Africa
| |
Collapse
|
3
|
Zhang W, Wang Y, Liu Y, Liu C, Wang Y, He L, Cheng X, Peng Y, Xia L, Wu X, Wu J, Zhang Y, Sun L, Chen P, Li G, Tu Q, Liang J, Shang Y. NFIB facilitates replication licensing by acting as a genome organizer. Nat Commun 2023; 14:5076. [PMID: 37604829 PMCID: PMC10442334 DOI: 10.1038/s41467-023-40846-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 08/12/2023] [Indexed: 08/23/2023] Open
Abstract
The chromatin-based rule governing the selection and activation of replication origins in metazoans remains to be investigated. Here we report that NFIB, a member of Nuclear Factor I (NFI) family that was initially purified in host cells to promote adenoviral DNA replication but has since mainly been investigated in transcription regulation, is physically associated with the pre-replication complex (pre-RC) in mammalian cells. Genomic analyses reveal that NFIB facilitates the assembly of the pre-RC by increasing chromatin accessibility. Nucleosome binding and single-molecule magnetic tweezers shows that NFIB binds to and opens up nucleosomes. Transmission electron microscopy indicates that NFIB promotes nucleosome eviction on parental chromatin. NFIB deficiency leads to alterations of chromosome contacts/compartments in both G1 and S phase and affects the firing of a subset of origins at early-replication domains. Significantly, cancer-associated NFIB overexpression provokes gene duplication and genomic alterations recapitulating the genetic aberrance in clinical breast cancer and empowering cancer cells to dynamically evolve growth advantage and drug resistance. Together, these results point a role for NFIB in facilitating replication licensing by acting as a genome organizer, shedding new lights on the biological function of NFIB and on the replication origin selection in eukaryotes.
Collapse
Affiliation(s)
- Wenting Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Yue Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou, 311121, China
| | - Yongjie Liu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Cuifang Liu
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yizhou Wang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Lin He
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Xiao Cheng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Yani Peng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Lu Xia
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Xiaodi Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Jiajing Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Yu Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Luyang Sun
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Ping Chen
- Department of Immunology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China
| | - Guohong Li
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Qiang Tu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Jing Liang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China.
| | - Yongfeng Shang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China.
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou, 311121, China.
| |
Collapse
|
4
|
Zhu R, Pan X, Wang S, Qiu Z, Gu C, Yao X, Li W. Updated skin transcriptomic atlas depicted by reciprocal contribution of single-nucleus RNA sequencing and single-cell RNA sequencing. J Dermatol Sci 2023; 111:22-31. [PMID: 37407342 DOI: 10.1016/j.jdermsci.2023.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 06/13/2023] [Accepted: 06/17/2023] [Indexed: 07/07/2023]
Abstract
BACKGROUND Single-cell RNA sequencing (scRNA-seq) has advanced our understanding of skin biology, but its utility is restricted by the requirement of fresh samples, inadequate dissociation-induced cell loss or death, and activation during tissue digestion. Single-nucleus RNA sequencing (snRNA-seq) can use frozen, hard-to-dissociate materials, which might be a promising method to circumvent the limitations of scRNA-seq for the skin tissue. OBJECTIVE To profile skin cells using snRNA-seq in parallel with scRNA-seq. METHODS We performed snRNA-seq in parallel with scRNA-seq for the bisected skin sample of one person and integrated previously published scRNA-seq data for analysis. We comparatively analyzed the differences in cell proportions and gene expression between the two methods. The differentiation trajectories of keratinocytes and fibroblasts were analyzed by Slingshot analysis. RESULTS snRNA-seq was less susceptible to contamination from mitochondrial and ribosomal RNA, and exhibited a greater capacity to detect transcription factors. snRNA-seq identified more spatially and functionally relevant keratinocyte clusters that constitute cell trajectories with expected differentiation dynamics. Novel markers, e.g., LYPD3, EMP2, and CSTB, were revealed for different differentiation stages of keratinocytes, and NFIB and GRHL1 were identified as transcription factors involving in the proliferation and functional differentiation of keratinocytes. Fibroblasts were found in a state of activation in scRNA-seq. And scRNA-seq detected a greater number of immune cells. CONCLUSIONS We generated an updated atlas of the skin transcriptome based on the reciprocal contribution of scRNA-seq and snRNA-seq.
Collapse
Affiliation(s)
- Ronghui Zhu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai Institute of Dermatology, Shanghai, China
| | - Xiaoyu Pan
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai Institute of Dermatology, Shanghai, China
| | - Shangshang Wang
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai Institute of Dermatology, Shanghai, China
| | - Zhuoqiong Qiu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai Institute of Dermatology, Shanghai, China
| | - Chaoying Gu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai Institute of Dermatology, Shanghai, China
| | - Xu Yao
- Department of Allergy and Rheumatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Wei Li
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai Institute of Dermatology, Shanghai, China.
| |
Collapse
|
5
|
Chen Y, Kong C, Yang M, Liu Y, Han Z, Xu L, Zheng X, Ding Y, Yin Z, Zhang X. 2,5-Hexanedione Affects Ovarian Granulosa Cells in Swine by Regulating the CDKN1A Gene: A Transcriptome Analysis. Vet Sci 2023; 10:vetsci10030201. [PMID: 36977240 PMCID: PMC10058995 DOI: 10.3390/vetsci10030201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/27/2023] [Accepted: 03/02/2023] [Indexed: 03/09/2023] Open
Abstract
N-hexane, a common industrial organic solvent, causes multiple organ damage owing to its metabolite, 2,5-hexanedione (2,5-HD). To identify and evaluate the effects of 2,5-HD on sows’ reproductive performance, we used porcine ovarian granulosa cells (pGCs) as a vehicle and carried out cell morphology and transcriptome analyses. 2,5-HD has the potential to inhibit the proliferation of pGCs and induce morphological changes and apoptosis depending on the dose. RNA-seq analyses identified 4817 differentially expressed genes (DEGs), with 2394 down-regulated and 2423 up-regulated following 2,5-HD exposure treatment. The DEG, cyclin-dependent kinase inhibitor 1A (CDKN1A), according to the Kyoto Encyclopedia of Genes and Genomes enrichment analysis, was significantly enriched in the p53 signaling pathway. Thus, we evaluated its function in pGC apoptosis in vitro. Then, we knocked down the CDKN1A gene in the pGCs to identify its effects on pGCs. Its knockdown decreased pGC apoptosis, with significantly fewer cells in the G1 phase (p < 0.05) and very significantly more cells in the S phase (p < 0.01). Herein, we revealed novel candidate genes that influence pGCs apoptosis and cell cycle and provided new insights into the role of CDKN1A in pGCs during apoptosis and cell cycle arrest.
Collapse
Affiliation(s)
- Yige Chen
- Anhui Provincial Laboratory of Local Animal Genetic Resource Conservation and Bio-Breeding, College of Animal Science and Technology, Anhui Agricultural University, No. 130, West Changjiang Road, Hefei 230036, China
| | - Chengcheng Kong
- Anhui Provincial Laboratory of Local Animal Genetic Resource Conservation and Bio-Breeding, College of Animal Science and Technology, Anhui Agricultural University, No. 130, West Changjiang Road, Hefei 230036, China
| | - Min Yang
- Anhui Provincial Laboratory of Local Animal Genetic Resource Conservation and Bio-Breeding, College of Animal Science and Technology, Anhui Agricultural University, No. 130, West Changjiang Road, Hefei 230036, China
- Anhui Province Key Laboratory of Aquaculture & Stock Enhancement, Fishery Institute of Anhui Academy of Agricultural Sciences, Hefei 230031, China
| | - Yangguang Liu
- Anhui Provincial Laboratory of Local Animal Genetic Resource Conservation and Bio-Breeding, College of Animal Science and Technology, Anhui Agricultural University, No. 130, West Changjiang Road, Hefei 230036, China
| | - Zheng Han
- Anhui Provincial Laboratory of Local Animal Genetic Resource Conservation and Bio-Breeding, College of Animal Science and Technology, Anhui Agricultural University, No. 130, West Changjiang Road, Hefei 230036, China
| | - Liming Xu
- Anhui Provincial Laboratory of Local Animal Genetic Resource Conservation and Bio-Breeding, College of Animal Science and Technology, Anhui Agricultural University, No. 130, West Changjiang Road, Hefei 230036, China
| | - Xianrui Zheng
- Anhui Provincial Laboratory of Local Animal Genetic Resource Conservation and Bio-Breeding, College of Animal Science and Technology, Anhui Agricultural University, No. 130, West Changjiang Road, Hefei 230036, China
| | - Yueyun Ding
- Anhui Provincial Laboratory of Local Animal Genetic Resource Conservation and Bio-Breeding, College of Animal Science and Technology, Anhui Agricultural University, No. 130, West Changjiang Road, Hefei 230036, China
| | - Zongjun Yin
- Anhui Provincial Laboratory of Local Animal Genetic Resource Conservation and Bio-Breeding, College of Animal Science and Technology, Anhui Agricultural University, No. 130, West Changjiang Road, Hefei 230036, China
- Correspondence: (Z.Y.); (X.Z.); Tel.: +86-13866191465 (Z.Y.); +86-15055138374 (X.Z.); Fax: +86-551-65787303 (Z.Y. & X.Z.)
| | - Xiaodong Zhang
- Anhui Provincial Laboratory of Local Animal Genetic Resource Conservation and Bio-Breeding, College of Animal Science and Technology, Anhui Agricultural University, No. 130, West Changjiang Road, Hefei 230036, China
- Correspondence: (Z.Y.); (X.Z.); Tel.: +86-13866191465 (Z.Y.); +86-15055138374 (X.Z.); Fax: +86-551-65787303 (Z.Y. & X.Z.)
| |
Collapse
|
6
|
Zhao W, Chen C, Zhou J, Chen X, Cai K, Shen M, Chen X, Jiang L, Wang G. Inhibition of Autophagy Promotes the Anti-Tumor Effect of Metformin in Oral Squamous Cell Carcinoma. Cancers (Basel) 2022; 14:cancers14174185. [PMID: 36077722 PMCID: PMC9454503 DOI: 10.3390/cancers14174185] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 08/25/2022] [Accepted: 08/26/2022] [Indexed: 11/16/2022] Open
Abstract
Oral Squamous Cell Carcinoma (OSCC) is the most common malignant tumor in the head and neck. Due to its high malignancy and easy recurrence, the five-year survival rate is only 50–60%. Currently, commonly used chemotherapy drugs for OSCC include cisplatin, paclitaxel, and fluorouracil, which are highly cytotoxic and cause drug resistance in patients. Therefore, a safe and effective treatment strategy for OSCC is urgent. To address this issue, our study investigated the anti-tumor activity of metformin (the first-line diabetes drug) in OSCC. We found that metformin could inhibit OSCC cell proliferation by promoting apoptosis and blocking the cell cycle in G1 phase. Additionally, we also found that metformin could induce protective autophagy of OSCC cells. After inhibiting autophagy with hydroxychloroquine (HCQ), the metformin-induced apoptosis was enhanced. In vitro, metformin inhibited the growth of subcutaneous xenograft tumor in nude mice and HCQ enhanced this effect of metformin. Therefore, metformin combined with HCQ may become a safe and effective treatment strategy for OSCC.
Collapse
Affiliation(s)
- Wei Zhao
- Department of Stomatology, Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Chen Chen
- Department of Stomatology, Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Jianjun Zhou
- Department of Stomatology, 900 Hospital of the Joint Logistics Team, Fuzhou 350025, China
| | - Xiaoqing Chen
- Department of Stomatology, Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Kuan Cai
- Department of Stomatology, Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Miaomiao Shen
- Department of Stomatology, Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Xuan Chen
- Department of Stomatology, Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Lei Jiang
- Department of Stomatology, Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Guodong Wang
- Department of Stomatology, Changzheng Hospital, Naval Medical University, Shanghai 200003, China
- Correspondence:
| |
Collapse
|
7
|
Uluca B, Lektemur Esen C, Saritas Erdogan S, Kumbasar A. NFI transcriptionally represses CDON and is required for SH-SY5Y cell survival. BIOCHIMICA ET BIOPHYSICA ACTA. GENE REGULATORY MECHANISMS 2022; 1865:194798. [PMID: 35151899 DOI: 10.1016/j.bbagrm.2022.194798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 01/14/2022] [Accepted: 01/30/2022] [Indexed: 06/14/2023]
Abstract
Nuclear Factor One (NFI) family of transcription factors regulate proliferation and multiple aspects of differentiation, playing analogous roles in embryonic development and various types of cancer. While all NFI family members are expressed in the developing brain and are involved in progression of brain cancers, their role in neuroblastoma has not been studied. Here we show that NFIB is required for the survival and proliferation of SH-SY5Y neuroblastoma cells, assessed by viability and colony formation assays. Cdon, an Ig superfamily member, is a SHH dependence receptor that acts as a tumor suppressor in neuroblastoma. In the absence of NFI, Cdon is upregulated in the developing mouse brain, however the mechanisms by which its transcription is regulated remains unknown. We report CDON as a downstream target of NFIs in SH-SY5Y cells. There are three putative NFI binding sites within the one kb CDON promoter, two of which are occupied by NFIs in SH-SY5Y cells and human neural stem cells. In dual-luciferase assays, Nfib directly represses CDON proximal promoter activity. Moreover, silencing NFIB leads to upregulation of CDON in SH-SY5Y cells, however, decreased cell proliferation in NFIB silenced cells could not be rescued by concomitantly silencing CDON, suggesting other molecular players are involved. For instance, p21, an NFI target in glioblastoma and breast cancer cells, is also upregulated upon NFIB knock-down. We propose that NFIB is indispensable for SH-SY5Y cells which may involve regulation of apoptosis inducer proteins CDON and p21.
Collapse
Affiliation(s)
- Betül Uluca
- Department of Molecular Biology and Genetics, Istanbul Technical University, Maslak, Istanbul 34469, Turkey; Department of Molecular Biotechnology, Turkish-German University, Beykoz, Istanbul 34820, Turkey
| | - Cemre Lektemur Esen
- Department of Molecular Biology and Genetics, Istanbul Technical University, Maslak, Istanbul 34469, Turkey
| | - Sinem Saritas Erdogan
- Department of Molecular Biology and Genetics, Istanbul Technical University, Maslak, Istanbul 34469, Turkey
| | - Asli Kumbasar
- Department of Molecular Biology and Genetics, Istanbul Technical University, Maslak, Istanbul 34469, Turkey.
| |
Collapse
|
8
|
Sulistyowati I, Sukpaita T, Limjeerajarus CN, Ampornaramveth RS. Hydroxamate-Based Histone Deacetylase Inhibitors as Potential Mediators to Induce Dentine Regeneration by Human Dental Pulp Cell. FRONTIERS IN DENTAL MEDICINE 2021. [DOI: 10.3389/fdmed.2021.765462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Human dental pulp cells (hDPCs) have shown their plasticity when treated with the hydroxamate-based histone deacetylase (HDAC) inhibitor members, Trichostatin A (TSA), and suberoylanilide hydroxamic acid (SAHA). However, a comparison of their potency to stimulate odontoblast-like differentiation and mineralization has not been reported. The aim of our study was to confirm and compare these TSA and SAHA effects. Primary hDPCs cultured with/without various TSA or SAHA concentrations were evaluated using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT), ALP activity, alizarin red staining, and scratch wound healing assays. The inhibitory effect of TSA and SAHA on inhibiting the activity of HDAC was evaluated by HDAC activity assay. Odontoblast-related gene expression was determined using RT-qPCR. The MTT assay indicated that TSA or SAHA did not affect hDPC viability. TSA or SAHA treatment-induced odontoblast-like differentiation as evidenced by a significant increase in alkaline phosphatase activity and mineral deposition after 400 nM TSA or 1 μM SAHA treatment. A significant increase in nuclear factor I C, kruppel like factor 4, dentin matrix acidic phosphoprotein 1, dentin sialophosphoprotein, collagen type I alpha 1 chain, alkaline phosphatase (ALPL), integrin-binding sialoprotein, bone gamma-carboxyglutamate protein, vascular endothelial growth factor A, and cyclin-dependent kinase inhibitor 1A gene expression analyzed by RT-qPCR, at 24, 72 h, 7, and 10 days of treatment. The activity of HDAC in hDPCs culture was significantly inhibited after 72 h TSA and SAHA treatment. The scratch wound healing assay displayed enhanced cell migration at 72 h after TSA or SAHA treatment. Our findings demonstrated that TSA and SAHA have similar stimulatory effects in inducing HDPC odontogenic differentiation and mineralization and propose another potential use of TSA and SAHA to promote dentin regeneration.
Collapse
|
9
|
Lee DS, Roh SY, Choi H, Park JC. NFI-C Is Required for Epiphyseal Chondrocyte Proliferation during Postnatal Cartilage Development. Mol Cells 2020; 43:739-748. [PMID: 32759468 PMCID: PMC7468589 DOI: 10.14348/molcells.2020.2272] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 07/01/2020] [Accepted: 07/26/2020] [Indexed: 12/24/2022] Open
Abstract
Stringent regulation of the chondrocyte cell cycle is required for endochondral bone formation. During the longitudinal growth of long bones, mesenchymal stem cells condense and differentiate into chondrocytes. Epiphyseal chondrocytes sequentially differentiate to form growth- plate cartilage, which is subsequently replaced with bone. Although the importance of nuclear factor 1C (Nfic) in hard tissue formation has been extensively studied, knowledge regarding its biological roles and molecular mechanisms in this process remains insufficient. Herein, we demonstrated that Nfic deficiency affects femoral growth-plate formation. Chondrocyte proliferation was downregulated and the number of apoptotic cell was increased in the growth plates of Nfic-/- mice. Further, the expression of the cell cycle inhibitor p21 was upregulated in the primary chondrocytes of Nfic-/- mice, whereas that of cyclin D1 was downregulated. Our findings suggest that Nfic may contribute to postnatal chondrocyte proliferation by inhibiting p21 expression and by increasing the stability of cyclin D1 protein.
Collapse
Affiliation(s)
- Dong-Seol Lee
- Laboratory for the Study of Regenerative Dental Medicine, Department of Oral Histology-Developmental Biology & Dental Research Institute, School of Dentistry, Seoul National University, Seoul 08826, Korea
- These authors contributed equally to this work
| | - Song Yi Roh
- Laboratory for the Study of Regenerative Dental Medicine, Department of Oral Histology-Developmental Biology & Dental Research Institute, School of Dentistry, Seoul National University, Seoul 08826, Korea
- These authors contributed equally to this work
| | - Hojae Choi
- Laboratory for the Study of Regenerative Dental Medicine, Department of Oral Histology-Developmental Biology & Dental Research Institute, School of Dentistry, Seoul National University, Seoul 08826, Korea
- Present address: Postgraduate Orthodontic Program, Arizona School of Dentistry & Oral Health, A.T. Still University, Mesa, AZ 8506, USA
| | - Joo-Cheol Park
- Laboratory for the Study of Regenerative Dental Medicine, Department of Oral Histology-Developmental Biology & Dental Research Institute, School of Dentistry, Seoul National University, Seoul 08826, Korea
| |
Collapse
|
10
|
Zeng J, Zhang H, Liu Y, Sun W, Yi D, Zhu L, Zhang Y, Pan X, Chen Y, Zhou Y, Bian G, Lai M, Zhou Q, Liu J, Chen B, Ma F. Overexpression of p21 Has Inhibitory Effect on Human Hematopoiesis by Blocking Generation of CD43+ Cells via Cell-Cycle Regulation. Int J Stem Cells 2020; 13:202-211. [PMID: 32587134 PMCID: PMC7378898 DOI: 10.15283/ijsc20033] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 03/07/2020] [Accepted: 05/15/2020] [Indexed: 12/12/2022] Open
Abstract
Background and Objectives p21, an important member of the Cip/Kip family, is involved in inhibitory effects of RUNX1b overexpression during the early stage of human hematopoiesis. Methods and Results We established a human embryonic stem cell (hESC) line with inducible expression of p21 (p21/hESCs). Overexpression of p21 did not influence either mesoderm induction or emergence of CD34+ cells, but it significantly decreased the production of CD43+ cells and changed the expression profile of hematopoiesis-related factors, leading to the negative effects of p21 on hematopoiesis. Conclusions In RUNX1b/hESC co-cultures when RUNX1b was induced from D0, perturbation of the cell cycle caused by upregulation of p21 probably prevented the appearance of CD43+ cells, but not CD34+ cells. The mechanisms via which CD34+ cells are blocked by RUNX1b overexpression remain to be elucidated.
Collapse
Affiliation(s)
- Jiahui Zeng
- Research Center for Stem Cell Therapies, Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Chengdu, China
| | - Huifang Zhang
- Research Center for Stem Cell Therapies, Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Chengdu, China
| | - Yuanling Liu
- Research Center for Stem Cell Therapies, Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Chengdu, China
| | - Wencui Sun
- Research Center for Stem Cell Therapies, Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Chengdu, China
| | - Danying Yi
- Research Center for Stem Cell Therapies, Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Chengdu, China
| | - Lijiao Zhu
- Research Center for Stem Cell Therapies, Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Chengdu, China
| | - Yonggang Zhang
- Research Center for Stem Cell Therapies, Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Chengdu, China
| | - Xu Pan
- Research Center for Stem Cell Therapies, Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Chengdu, China
| | - Yijing Chen
- Research Center for Stem Cell Therapies, Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Chengdu, China
| | - Ya Zhou
- Research Center for Stem Cell Therapies, Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Chengdu, China
| | - Guohui Bian
- Research Center for Stem Cell Therapies, Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Chengdu, China
| | - Mowen Lai
- Research Center for Stem Cell Therapies, Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Chengdu, China
| | - Qiongxiu Zhou
- Research Center for Stem Cell Therapies, Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Chengdu, China
| | - Jiaxin Liu
- Research Center for Stem Cell Therapies, Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Chengdu, China
| | - Bo Chen
- Research Center for Stem Cell Therapies, Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Chengdu, China
| | - Feng Ma
- Research Center for Stem Cell Therapies, Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), Chengdu, China.,State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China.,State Key Laboratory of Experimental Hematology, CAMS & PUMC, Tianjin, China
| |
Collapse
|
11
|
Le-Bel G, Cortez Ghio S, Guérin LP, Bisson F, Germain L, Guérin SL. Irradiated Human Fibroblasts as a Substitute Feeder Layer to Irradiated Mouse 3T3 for the Culture of Human Corneal Epithelial Cells: Impact on the Stability of the Transcription Factors Sp1 and NFI. Int J Mol Sci 2019; 20:ijms20246296. [PMID: 31847118 PMCID: PMC6940969 DOI: 10.3390/ijms20246296] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 12/03/2019] [Accepted: 12/09/2019] [Indexed: 12/26/2022] Open
Abstract
Because of the worldwide shortage of graftable corneas, alternatives to restore visual impairments, such as the production of a functional human cornea by tissue engineering, have emerged. Self-renewal of the corneal epithelium through the maintenance of a sub-population of corneal stem cells is required to maintain the functionality of such a reconstructed cornea. We previously reported an association between stem cell differentiation and the level to which they express the transcription factors Sp1 and NFI. In this study, we investigated the impact of replacing irradiated 3T3 (i3T3) murine fibroblast feeder cells by irradiated human corneal fibroblasts (iHFL) on the expression of Sp1 and NFI and evaluated their contribution to the proliferative properties of human corneal epithelial cells (hCECs) in both monolayer cultures and human tissue engineered corneas (hTECs). hCECs co-cultured with iHFL could be maintained for up to two more passages than when they were grown with i3T3. Western Blot and electrophoretic mobility shift assays (EMSAs) revealed no significant difference in the feeder-layer dependent increase in Sp1 at both the protein and DNA binding level, respectively, between HCECs grown with either i3T3 or iHFL. On the other hand, a significant increase in the expression and DNA binding of NFI was observed at each subsequent passage when hCECs were co-cultured along with i3T3. These changes were found to result from an increased expression of the NFIA and NFIB isoforms in hCECs grown with i3T3. Exposure of hCECs to cycloheximide revealed an increased stability of NFIB that likely resulted from post-translational glycosylation of this protein when these cells were co-cultured with i3T3. In addition, iHFL were as efficient as i3T3 at preserving corneal, slow-cycling, epithelial stem cells in the basal epithelium of the reconstructed hTECs. Furthermore, we observed an increased expression of genes whose encoded products promote hCECs differentiation along several passages in hCECs co-cultured with either type of feeder layer. Therefore, the iHFL feeder layer appears to be the most effective at maintaining the proliferative properties of hCECs in culture most likely by preserving high levels of Sp1 and low levels of NFIB, which is known for its gene repressor and cell differentiation properties.
Collapse
Affiliation(s)
- Gaëtan Le-Bel
- Centre LOEX de l’Université Laval, Génie Tissulaire et Régénération, Centre de Recherche du CHU de Québec -Université Laval, Axe Médecine Régénératrice, Québec, QC G1V 0A6, Canada; (G.L.-B.); (S.C.G.); (L.-P.G.); (F.B.); (L.G.)
- Centre Universitaire d’Ophtalmologie (CUO)-Recherche, Centre de recherche FRQS du CHU de Québec, Axe Médecine Régénératrice, Québec, QC G1J 1Z4, Canada
- Département de Chirurgie, Faculté de médecine, Université Laval, Québec, QC G1V 0A6, Canada
| | - Sergio Cortez Ghio
- Centre LOEX de l’Université Laval, Génie Tissulaire et Régénération, Centre de Recherche du CHU de Québec -Université Laval, Axe Médecine Régénératrice, Québec, QC G1V 0A6, Canada; (G.L.-B.); (S.C.G.); (L.-P.G.); (F.B.); (L.G.)
- Département de Chirurgie, Faculté de médecine, Université Laval, Québec, QC G1V 0A6, Canada
| | - Louis-Philippe Guérin
- Centre LOEX de l’Université Laval, Génie Tissulaire et Régénération, Centre de Recherche du CHU de Québec -Université Laval, Axe Médecine Régénératrice, Québec, QC G1V 0A6, Canada; (G.L.-B.); (S.C.G.); (L.-P.G.); (F.B.); (L.G.)
- Département d’Ophtalmologie, Faculté de médecine, Université Laval, Québec, QC G1V 0A6, Canada
| | - Francis Bisson
- Centre LOEX de l’Université Laval, Génie Tissulaire et Régénération, Centre de Recherche du CHU de Québec -Université Laval, Axe Médecine Régénératrice, Québec, QC G1V 0A6, Canada; (G.L.-B.); (S.C.G.); (L.-P.G.); (F.B.); (L.G.)
- Département de Chirurgie, Faculté de médecine, Université Laval, Québec, QC G1V 0A6, Canada
| | - Lucie Germain
- Centre LOEX de l’Université Laval, Génie Tissulaire et Régénération, Centre de Recherche du CHU de Québec -Université Laval, Axe Médecine Régénératrice, Québec, QC G1V 0A6, Canada; (G.L.-B.); (S.C.G.); (L.-P.G.); (F.B.); (L.G.)
- Département de Chirurgie, Faculté de médecine, Université Laval, Québec, QC G1V 0A6, Canada
- Département d’Ophtalmologie, Faculté de médecine, Université Laval, Québec, QC G1V 0A6, Canada
| | - Sylvain L. Guérin
- Centre LOEX de l’Université Laval, Génie Tissulaire et Régénération, Centre de Recherche du CHU de Québec -Université Laval, Axe Médecine Régénératrice, Québec, QC G1V 0A6, Canada; (G.L.-B.); (S.C.G.); (L.-P.G.); (F.B.); (L.G.)
- Centre Universitaire d’Ophtalmologie (CUO)-Recherche, Centre de recherche FRQS du CHU de Québec, Axe Médecine Régénératrice, Québec, QC G1J 1Z4, Canada
- Département d’Ophtalmologie, Faculté de médecine, Université Laval, Québec, QC G1V 0A6, Canada
- Correspondence: ; Tel.: +1-418-682-7565
| |
Collapse
|
12
|
Fang Z, Sun Y, Zhang X, Wang G, Li Y, Wang Y, Zhang Z. Responses of HSP70 Gene to Vibrio parahaemolyticus Infection and Thermal Stress and Its Transcriptional Regulation Analysis in Haliotis diversicolor. Molecules 2019; 24:E162. [PMID: 30609869 PMCID: PMC6337134 DOI: 10.3390/molecules24010162] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 12/04/2018] [Accepted: 12/17/2018] [Indexed: 02/08/2023] Open
Abstract
Heat-shock protein 70 (HSP70) is a molecular chaperone that plays critical roles in cell protein folding and metabolism, which helps to protect cells from unfavorable environmental stress. Haliotis diversicolor is one of the most important economic breeding species in the coastal provinces of south China. To date, the expression and transcriptional regulation of HSP70 in Haliotis diversicolor (HdHSP70) has not been well characterized. In this study, the expression levels of HdHSP70 gene in different tissues and different stress conditions were detected. The results showed that the HdHSP70 gene was ubiquitously expressed in sampled tissues and was the highest in hepatopancreas, followed by hemocytes. In hepatopancreas and hemocytes, the HdHSP70 gene was significantly up-regulated by Vibrio parahaemolyticus infection, thermal stress, and combined stress (Vibrio parahaemolyticus infection and thermal stress combination), indicating that HdHSP70 is involved in the stress response and the regulation of innate immunity. Furthermore, a 2383 bp of 5'-flanking region sequence of the HdHSP70 gene was cloned, and it contains a presumed core promoter region, a CpG island, a (TG)39 simple sequence repeat (SSR), and many potential transcription factor binding sites. The activity of HdHSP70 promoter was evaluated by driving the expression of luciferase gene in HEK293FT cells. A series of experimental results indicated that the core promoter region is located between -189 bp and +46 bp, and high-temperature stress can increase the activity of HdHSP70 promoter. Sequence-consecutive deletions of the luciferase reporter gene in HEK293FT cells revealed two possible promoter activity regions. To further identify the binding site of the key transcription factor in the two regions, two expression vectors with site-directed mutation were constructed. The results showed that the transcriptional activity of NF-1 site-directed mutation was significantly increased (p < 0.05), whereas the transcriptional activity of NF-κB site-directed mutation was significantly reduced. These results suggest that NF-1 and NF-κB may be two important transcription factors that regulate the expression of HdHSP70 gene.
Collapse
Affiliation(s)
- Zhiqiang Fang
- Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture, Fisheries College, Jimei University, Xiamen 361021, China.
| | - Yulong Sun
- College of Animal Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China.
| | - Xin Zhang
- College of Animal Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China.
| | - Guodong Wang
- Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture, Fisheries College, Jimei University, Xiamen 361021, China.
| | - Yuting Li
- Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture, Fisheries College, Jimei University, Xiamen 361021, China.
| | - Yilei Wang
- Key Laboratory of Healthy Mariculture for the East China Sea, Ministry of Agriculture, Fisheries College, Jimei University, Xiamen 361021, China.
| | - Ziping Zhang
- College of Animal Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China.
| |
Collapse
|
13
|
Xie W, Stopsack KH, Drouin SJ, Fu H, Pomerantz MM, Mucci LA, Lee GSM, Kantoff PW. Association of genetic variation of the six gene prognostic model for castration-resistant prostate cancer with survival. Prostate 2019; 79:73-80. [PMID: 30141208 PMCID: PMC6476182 DOI: 10.1002/pros.23712] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 08/08/2018] [Indexed: 11/08/2022]
Abstract
BACKGROUND We previously identified a blood RNA transcript-based model consisting of six immune or inflammatory response genes (ABL2, SEMA4D, ITGAL, C1QA, TIMP1, and CDKN1A) that was prognostic for survival in cohorts of men with castration-resistant prostate cancer (CRPC). We investigated whether inherited variation in these six genes was associated with overall survival (OS) in men with CRPC. METHODS The test cohort comprised 600 patients diagnosed with CRPC between 1996 and 2011 at Dana-Farber Cancer Institute. Genotyping of 66 tagging single nucleotide polymorphisms (SNPs) spanning the six genes was performed on blood derived DNAs. For the top four SNPs (P < 0.05), validation was conducted in an independent cohort of 223 men diagnosed with CRPC between 2000 and 2014. Multivariable Cox regression adjusting for known prognostic factors estimated hazard ratios (HR) and 95% confidence intervals (CI) of the association of genetic variants with OS. RESULTS Two thirds of patients in both cohorts had metastases at CRPC diagnosis. Median OS from CRPC diagnosis was 3.6 (95%CI 3.3-4.0) years in the test cohort and 4.6 (95%CI 3.8-5.2) years in the validation cohort. Fifty-nine SNPs in Hardy-Weinberg equilibrium were analyzed. The major alleles of rs1318056 and rs1490311 in ABL2, and the minor alleles of rs2073917 and rs3764322 in ITGAL were associated with increased risk of death in the test cohort (adjusted-HRs 1.27-1.39; adjusted-p <0.05; false discovery rate <0.35). In the validation cohort, a similar association with OS was observed for rs1318056 in ABL2 (adjusted-HR 1.44; 95%CI 0.89-2.34) and rs2073917 in ITGAL (adjusted-HR 1.41; 95%CI 0.82-2.42). The associations did not reach statistical significance most likely due to the small sample size of the validation cohort (adjusted-p = 0.142 and 0.209, respectively). Additional eQTL analysis indicated that minor alleles of rs1318056 and rs1490311 in ABL2 are associated with a lower ABL2 expression in blood. CONCLUSIONS These findings corroborate our initial work on the RNA expression of genes involved in immunity and inflammation from blood and clinical outcome and suggest that germline polymorphisms in ABL2 and ITGAL may be associated with the risk of death in men with CRPC. Further studies are needed to validate these findings and to explore their functional mechanisms.
Collapse
Affiliation(s)
- Wanling Xie
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute and Harvard Medical School, 450 Brookline Ave., Boston, MA 02215
| | - Konrad H. Stopsack
- Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065
| | - Sarah J Drouin
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, 450 Brookline Ave., Boston, MA 02215
| | - Henry Fu
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, 450 Brookline Ave., Boston, MA 02215
| | - Mark M. Pomerantz
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, 450 Brookline Ave., Boston, MA 02215
| | - Lorelei A. Mucci
- Harvard T. H Chan Harvard School of Public Health, 677 Huntington Avenue, Boston, MA 02215
| | - Gwo-Shu Mary Lee
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, 450 Brookline Ave., Boston, MA 02215
- Correspondence: Philip W. Kantoff, Phone: 212-639-5851; Fax: 929-321-5023; . Gwo-Shu Mary Lee, Phone: 617-632-5088;
| | - Philip W Kantoff
- Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065
- Correspondence: Philip W. Kantoff, Phone: 212-639-5851; Fax: 929-321-5023; . Gwo-Shu Mary Lee, Phone: 617-632-5088;
| |
Collapse
|
14
|
Liu RZ, Vo TM, Jain S, Choi WS, Garcia E, Monckton EA, Mackey JR, Godbout R. NFIB promotes cell survival by directly suppressing p21 transcription in TP53-mutated triple-negative breast cancer. J Pathol 2018; 247:186-198. [PMID: 30350349 DOI: 10.1002/path.5182] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 09/21/2018] [Accepted: 10/10/2018] [Indexed: 12/27/2022]
Abstract
Triple-negative breast cancer (TNBC) is an aggressive breast cancer subtype with limited treatment options and poor prognosis. There is an urgent need to identify and understand the key factors and signalling pathways driving TNBC tumour progression, relapse, and treatment resistance. In this study, we report that gene copy numbers and expression levels of nuclear factor IB (NFIB), a recently identified oncogene in small cell lung cancer, are preferentially increased in TNBC compared to other breast cancer subtypes. Furthermore, increased levels of NFIB are significantly associated with high tumour grade, poor prognosis, and reduced chemotherapy response. Concurrent TP53 mutations and NFIB overexpression (z-scores > 0) were observed in 77.9% of TNBCs, in contrast to 28.5% in non-TNBCs. Depletion of NFIB in TP53-mutated TNBC cell lines promotes cell death, cell cycle arrest, and enhances sensitivity to docetaxel, a first-line chemotherapeutic drug in breast cancer treatment. Importantly, these alterations in growth properties were accompanied by induction of CDKN1A, the gene encoding p21, a downstream effector of p53. We show that NFIB directly interacts with the CDKN1A promoter in TNBC cells. Furthermore, knockdown of combined p21 and NFIB reverses the docetaxel-induced cell growth inhibition observed upon NFIB knockdown, indicating that NFIB's effect on chemotherapeutic drug response is mediated through p21. Our results indicate that NFIB is an important TNBC factor that drives tumour cell growth and drug resistance, leading to poor clinical outcomes. Thus, targeting NFIB in TP53-mutated TNBC may reverse oncogenic properties associated with mutant p53 by restoring p21 activity. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Rong-Zong Liu
- Department of Oncology, University of Alberta, Cross Cancer Institute, Edmonton, Canada
| | - The M Vo
- Department of Oncology, University of Alberta, Cross Cancer Institute, Edmonton, Canada
| | - Saket Jain
- Department of Oncology, University of Alberta, Cross Cancer Institute, Edmonton, Canada
| | - Won-Shik Choi
- Department of Oncology, University of Alberta, Cross Cancer Institute, Edmonton, Canada
| | - Elizabeth Garcia
- Department of Oncology, University of Alberta, Cross Cancer Institute, Edmonton, Canada
| | - Elizabeth A Monckton
- Department of Oncology, University of Alberta, Cross Cancer Institute, Edmonton, Canada
| | - John R Mackey
- Department of Oncology, University of Alberta, Cross Cancer Institute, Edmonton, Canada
| | - Roseline Godbout
- Department of Oncology, University of Alberta, Cross Cancer Institute, Edmonton, Canada
| |
Collapse
|
15
|
Le-Bel G, Giasson CJ, Deschambeault A, Carrier P, Germain L, Guérin SL. The presence of a feeder layer improves human corneal endothelial cell proliferation by altering the expression of the transcription factors Sp1 and NFI. Exp Eye Res 2018; 176:161-173. [PMID: 30003884 DOI: 10.1016/j.exer.2018.07.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 06/13/2018] [Accepted: 07/06/2018] [Indexed: 12/13/2022]
Abstract
Based on the use of tissue-cultured human corneal endothelial cells (HCECs), cell therapy is a very promising avenue in the treatment of corneal endothelial pathologies such as Fuchs' dystrophy, and post-surgical corneal edema. However, once in culture, HCECs rapidly lose their phenotypic and physiological characteristics, and are therefore unsuitable for the reconstruction of a functional endothelial monolayer. Expression of NFI, a transcription factor that can either function as an activator or a repressor of gene transcription, has never been examined in endothelial cells. The present study therefore aimed to determine the impact of a non-proliferating, lethally irradiated i3T3 feeder layer on the maintenance of HCEC's morphological characteristics, and both the expression and stability of Sp1 (a strong transcriptional activator) and NFI in such cells. The typical morphology of endothelial cells was best maintained when 8 × 103/cm2 HCECs were co-cultured in the presence of 2 × 104 cells/cm2 i3T3. HCECs were found to express both Sp1 and NFI in vitro. Also, the presence of i3T3 led to higher levels of Sp1 and NFI in HCECs, with a concomitant increase in their DNA binding levels (assessed by electrophoretic mobility shift assays (EMSA)). Specifically, i3T3 increased the expression of the NFIA, NFIB and NFIC isoforms, without a noticeable increase in their mRNAs (as revealed by gene profiling on microarray). Gene profiling analysis also identified a few feeder layer-dependent, differentially regulated genes whose protein products may contribute to improving the properties of HCECs in culture. Therefore, co-culturing HCECs with an i3T3 feeder layer clearly improves their morphological characteristics by maintaining stable levels of Sp1 and NFI in cell culture.
Collapse
Affiliation(s)
- Gaëtan Le-Bel
- CUO-Recherche, Médecine Régénératrice - Centre de recherche FRQS du CHU de Québec-Université Laval, Québec, Canada; Centre de Recherche en Organogénèse expérimentale de l'Université Laval/LOEX, Québec, QC, Canada; Département d'Ophtalmologie, Faculté de médecine, Université Laval, Québec, QC, Canada; Département de Chirurgie, Faculté de médecine, Université Laval, Québec, QC, Canada and
| | - Claude J Giasson
- CUO-Recherche, Médecine Régénératrice - Centre de recherche FRQS du CHU de Québec-Université Laval, Québec, Canada; Centre de Recherche en Organogénèse expérimentale de l'Université Laval/LOEX, Québec, QC, Canada; École d'Optométrie, Université de Montréal, Montréal, Québec, Canada
| | - Alexandre Deschambeault
- CUO-Recherche, Médecine Régénératrice - Centre de recherche FRQS du CHU de Québec-Université Laval, Québec, Canada; Centre de Recherche en Organogénèse expérimentale de l'Université Laval/LOEX, Québec, QC, Canada; Département de Chirurgie, Faculté de médecine, Université Laval, Québec, QC, Canada and
| | - Patrick Carrier
- CUO-Recherche, Médecine Régénératrice - Centre de recherche FRQS du CHU de Québec-Université Laval, Québec, Canada; Centre de Recherche en Organogénèse expérimentale de l'Université Laval/LOEX, Québec, QC, Canada; Département de Chirurgie, Faculté de médecine, Université Laval, Québec, QC, Canada and
| | - Lucie Germain
- CUO-Recherche, Médecine Régénératrice - Centre de recherche FRQS du CHU de Québec-Université Laval, Québec, Canada; Centre de Recherche en Organogénèse expérimentale de l'Université Laval/LOEX, Québec, QC, Canada; Département d'Ophtalmologie, Faculté de médecine, Université Laval, Québec, QC, Canada; Département de Chirurgie, Faculté de médecine, Université Laval, Québec, QC, Canada and
| | - Sylvain L Guérin
- CUO-Recherche, Médecine Régénératrice - Centre de recherche FRQS du CHU de Québec-Université Laval, Québec, Canada; Centre de Recherche en Organogénèse expérimentale de l'Université Laval/LOEX, Québec, QC, Canada; Département d'Ophtalmologie, Faculté de médecine, Université Laval, Québec, QC, Canada.
| |
Collapse
|
16
|
Brun M, Jain S, Monckton EA, Godbout R. Nuclear Factor I Represses the Notch Effector HEY1 in Glioblastoma. Neoplasia 2018; 20:1023-1037. [PMID: 30195713 PMCID: PMC6138789 DOI: 10.1016/j.neo.2018.08.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 08/17/2018] [Accepted: 08/20/2018] [Indexed: 01/16/2023] Open
Abstract
Glioblastomas (GBMs) are highly aggressive brain tumors with a dismal prognosis. Nuclear factor I (NFI) is a family of transcription factors that controls glial cell differentiation in the developing central nervous system. NFIs have previously been shown to regulate the expression of astrocyte markers such as glial fibrillary acidic protein (GFAP) in both normal brain and GBM cells. We used chromatin immunoprecipitation (ChIP)–on-chip to identify additional NFI targets in GBM cells. Analysis of our ChIP data revealed ~400 putative NFI target genes including an effector of the Notch signaling pathway, HEY1, implicated in the maintenance of neural stem cells. All four NFIs (NFIA, NFIB, NFIC, and NFIX) bind to NFI recognition sites located within 1 kb upstream of the HEY1 transcription site. We further showed that NFI negatively regulates HEY1 expression, with knockdown of all four NFIs in GBM cells resulting in increased HEY1 RNA levels. HEY1 knockdown in GBM cells decreased cell proliferation, increased cell migration, and decreased neurosphere formation. Finally, we found a general correlation between elevated levels of HEY1 and expression of the brain neural stem/progenitor cell marker B-FABP in GBM cell lines. Knockdown of HEY1 resulted in an increase in the RNA levels of the GFAP astrocyte differentiation marker. Overall, our data indicate that HEY1 is negatively regulated by NFI family members and is associated with increased proliferation, decreased migration, and increased stem cell properties in GBM cells.
Collapse
Affiliation(s)
- Miranda Brun
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada, T6G 1Z2
| | - Saket Jain
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada, T6G 1Z2
| | - Elizabeth A Monckton
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada, T6G 1Z2
| | - Roseline Godbout
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada, T6G 1Z2.
| |
Collapse
|
17
|
Dai J, Kumbhare A, Williams DA, Youssef D, Yao ZQ, McCall CE, El Gazzar M. Nfia deletion in myeloid cells blocks expansion of myeloid-derived suppressor cells during sepsis. Innate Immun 2017; 24:54-65. [PMID: 29172874 PMCID: PMC6240914 DOI: 10.1177/1753425917742956] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Sepsis-induced immunosuppression increases the risk of chronic infection and
reduces survival. Myeloid-derived suppressor cells (MDSCs) expand in the bone
marrow and spleen during murine polymicrobial sepsis, contributing to
immunosuppression. A better understanding of molecular controls of MDSC
production is needed to identify treatment targets. We previously reported that
miR-21 and miR-181b couple with transcription factor NFI-A to induce MDSCs
during murine sepsis. Here, we expand upon these observations by showing that
conditional deletion of the Nfia gene in the
myeloid lineage precludes MDSC development. NFI-A-deficient
Gr1+CD11b+ myeloid cells are not immunosuppressive and
differentiate normally into macrophages and dendritic cells. In contrast,
ectopically expressed NFI-A prevents differentiation of these immature
Gr1+CD11b+ cells, while converting them into MDSCs. In
addition, NFI-A-deficient Gr1+CD11b+ cells decreased, and
cells transfected with NFI-A increase expression of miR-21 and miR181b. Our
results support a myeloid cell loop in which NFI-A and miR-21 and miR-181b
sustain Gr1+CD11b+ MDSC-dependent immunosuppression during
sepsis.
Collapse
Affiliation(s)
- Jun Dai
- 1 Department of Internal Medicine, 12324 East Tennessee State University College of Medicine , Johnson City, TN, USA
| | - Ajinkya Kumbhare
- 1 Department of Internal Medicine, 12324 East Tennessee State University College of Medicine , Johnson City, TN, USA
| | - Danielle A Williams
- 2 Department of Health Sciences, 144478 East Tennessee State University College of Public Health , Johnson City, TN, USA
| | - Dima Youssef
- 1 Department of Internal Medicine, 12324 East Tennessee State University College of Medicine , Johnson City, TN, USA
| | - Zhi Q Yao
- 1 Department of Internal Medicine, 12324 East Tennessee State University College of Medicine , Johnson City, TN, USA
| | - Charles E McCall
- 3 Department of Internal Medicine, Section of Molecular Medicine, 12279 Wake Forest University School of Medicine , Winston-Salem, NC, USA
| | - Mohamed El Gazzar
- 1 Department of Internal Medicine, 12324 East Tennessee State University College of Medicine , Johnson City, TN, USA
| |
Collapse
|
18
|
Han X, Li H, Zhang Y, Qin J, Yang Q, Wang L, Yuan M, Xia C. Brain lipid-binding protein promotes proliferation and modulates cell cycle in C6 rat glioma cells. Int J Oncol 2017; 51:1439-1448. [PMID: 29048614 PMCID: PMC5642387 DOI: 10.3892/ijo.2017.4132] [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: 04/11/2017] [Accepted: 09/18/2017] [Indexed: 12/13/2022] Open
Abstract
Gliomas are the most common primary brain tumors affecting adults. Four grades of gliomas have been identified, namely, grades I-IV. Brain lipid-binding protein (BLBP), which functions in the intracellular transport of fatty acids, is expressed in all grades of human gliomas. The glioma cells that are cultured in vitro are grouped into the BLBP-positive and BLBP-negative cell lines. In the present study, we found that C6 rat glioma cells was a distinct type of BLBP-negative cell line. Our results confirmed that in the C6 cells, the expression of exogenous BLBP increased the proliferation and percentage of cells in the S phase, in the culture medium containing 10 or 1% FBS. Moreover, exogenous BLBP was found to downregulate the tumor suppressors p21 and p16 in the 1% FBS culture medium, but only p21 in the 10% FBS culture medium. The results of the xenograft model assay showed that exogenous BLBP also stimulated tumor formation and downregulated p21 and p16. In conclusion, our study demonstrated that exogenous BLBP promoted proliferation of the C6 cells in vitro and facilitated tumor formation in vivo. Therefore, BLBP expression in glioma cells may promote cell growth by inhibiting the tumor suppressors.
Collapse
Affiliation(s)
- Xiao Han
- Department of Anatomy and Cytoneurobiology Unit, Medical College of Soochow University, Suzhou, Jiangsu 215123, P.R. China
| | - Haoming Li
- Department of Human Anatomy, Medical School of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Ye Zhang
- Department of Anatomy and Cytoneurobiology Unit, Medical College of Soochow University, Suzhou, Jiangsu 215123, P.R. China
| | - Jianbing Qin
- Department of Human Anatomy, Medical School of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Qingqing Yang
- Department of Medicine, Xinglin College of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Lu Wang
- Department of Human Anatomy, Medical School of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Mingjie Yuan
- Department of Human Anatomy, Medical School of Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Chunlin Xia
- Department of Anatomy and Cytoneurobiology Unit, Medical College of Soochow University, Suzhou, Jiangsu 215123, P.R. China
| |
Collapse
|
19
|
Lim JC, Ko KI, Mattos M, Fang M, Zhang C, Feinberg D, Sindi H, Li S, Alblowi J, Kayal RA, Einhorn TA, Gerstenfeld LC, Graves DT. TNFα contributes to diabetes impaired angiogenesis in fracture healing. Bone 2017; 99:26-38. [PMID: 28285015 PMCID: PMC5563392 DOI: 10.1016/j.bone.2017.02.014] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 01/05/2017] [Accepted: 02/26/2017] [Indexed: 02/07/2023]
Abstract
Diabetes increases the likelihood of fracture, interferes with fracture healing and impairs angiogenesis. The latter may be significant due to the critical nature of angiogenesis in fracture healing. Although it is known that diabetes interferes with angiogenesis the mechanisms remain poorly defined. We examined fracture healing in normoglycemic and streptozotocin-induced diabetic mice and quantified the degree of angiogenesis with antibodies to three different vascular markers, CD34, CD31 and Factor VIII. The role of diabetes-enhanced inflammation was investigated by treatment of the TNFα-specific inhibitor, pegsunercept starting 10days after induction of fractures. Diabetes decreased both angiogenesis and VEGFA expression by chondrocytes. The reduced angiogenesis and VEGFA expression in diabetic fractures was rescued by specific inhibition of TNF in vivo. In addition, the TNF inhibitor rescued the negative effect of diabetes on endothelial cell proliferation and endothelial cell apoptosis. The effect of TNFα in vitro was enhanced by high glucose and an advanced glycation endproduct to impair microvascular endothelial cell proliferation and tube formation and to stimulate apoptosis. The effect of TNF, high glucose and an AGE was mediated by the transcription factor FOXO1, which increased expression of p21 and caspase-3. These studies indicate that inflammation plays a major role in diabetes-impaired angiogenesis in endochondral bone formation through its effect on microvascular endothelial cells and FOXO1.
Collapse
Affiliation(s)
- Jason C Lim
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kang I Ko
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Marcelo Mattos
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Miao Fang
- Department of Endocrinology, Shanxi Province People's Hospital, Shanxi Province, China
| | - Citong Zhang
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Implantology, School of Stomatology, Jilin University, Changchun 130021, China
| | - Daniel Feinberg
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hisham Sindi
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shuai Li
- Department of Implant Dentistry, Peking University, School and Hospital of Stomatology, Beijing, China
| | - Jazia Alblowi
- Department of Oral Basic and Clinical Sciences, Faculty of Dentistry, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Rayyan A Kayal
- Department of Oral Basic and Clinical Sciences, Faculty of Dentistry, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Thomas A Einhorn
- Department of Orthopedic Surgery, School of Medicine, Boston University, Boston, MA 02118, USA
| | - Louis C Gerstenfeld
- Department of Orthopedic Surgery, School of Medicine, Boston University, Boston, MA 02118, USA
| | - Dana T Graves
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
20
|
Roh SY, Park JC. The role of nuclear factor I-C in tooth and bone development. J Korean Assoc Oral Maxillofac Surg 2017; 43:63-69. [PMID: 28462188 PMCID: PMC5410429 DOI: 10.5125/jkaoms.2017.43.2.63] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 04/06/2017] [Indexed: 01/15/2023] Open
Abstract
Nuclear factor I-C (NFI-C) plays a pivotal role in various cellular processes such as odontoblast and osteoblast differentiation. Nfic-deficient mice showed abnormal tooth and bone formation. The transplantation of Nfic-expressing mouse bone marrow stromal cells rescued the impaired bone formation in Nfic-/- mice. Studies suggest that NFI-C regulate osteogenesis and dentinogenesis in concert with several factors including transforming growth factor-β1, Krüppel-like factor 4, and β-catenin. This review will focus on the function of NFI-C during tooth and bone formation and on the relevant pathways that involve NFI-C.
Collapse
Affiliation(s)
- Song Yi Roh
- Department of Oral Histology-Developmental Biology and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, Korea
| | - Joo-Cheol Park
- Department of Oral Histology-Developmental Biology and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, Korea
| |
Collapse
|
21
|
Kanashiro-Takeuchi RM, Szalontay L, Schally AV, Takeuchi LM, Popovics P, Jaszberenyi M, Vidaurre I, Zarandi M, Cai RZ, Block NL, Hare JM, Rick FG. New therapeutic approach to heart failure due to myocardial infarction based on targeting growth hormone-releasing hormone receptor. Oncotarget 2016; 6:9728-39. [PMID: 25797248 PMCID: PMC4496393 DOI: 10.18632/oncotarget.3303] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 02/08/2015] [Indexed: 12/21/2022] Open
Abstract
Background We previously showed that growth hormone-releasing hormone (GHRH) agonists are cardioprotective following myocardial infarction (MI). Here, our aim was to evaluate the in vitro and in vivo activities of highly potent new GHRH agonists, and elucidate their mechanisms of action in promoting cardiac repair. Methods and Results H9c2 cells were cultured in serum-free medium, mimicking nutritional deprivation. GHRH agonists decreased calcium influx and significantly improved cell survival. Rats with cardiac infarction were treated with GHRH agonists or placebo for four weeks. MI size was reduced by selected GHRH agonists (JI-38, MR-356, MR-409); this accompanied an increased number of cardiac c-kit+ cells, cellular mitotic divisions, and vascular density. One week post-MI, MR-409 significantly reduced plasma levels of IL-2, IL-6, IL-10 and TNF-α compared to placebo. Gene expression studies revealed favorable outcomes of MR-409 treatment partially result from inhibitory activity on pro-apoptotic molecules and pro-fibrotic systems, and by elevation of bone morphogenetic proteins. Conclusions Treatment with GHRH agonists appears to reduce the inflammatory responses post-MI and may consequently improve mechanisms of healing and cardiac remod eling by regulating pathways involved in fibrosis, apoptosis and cardiac repair. Patients with cardiac dysfunction could benefit from treatment with novel GHRH agonists.
Collapse
Affiliation(s)
- Rosemeire M Kanashiro-Takeuchi
- Interdisciplinary Stem Cell Institute, University of Miami, Miller School of Medicine, Miami, Florida, United States of America.,Department of Molecular and Cellular Pharmacology, University of Miami, Miller School of Medicine, Florida, United States of America
| | - Luca Szalontay
- Veterans Affairs Medical Center and South Florida Veterans Affairs Foundation for Research and Education, Miami, Florida, United States of America
| | - Andrew V Schally
- Interdisciplinary Stem Cell Institute, University of Miami, Miller School of Medicine, Miami, Florida, United States of America.,Veterans Affairs Medical Center and South Florida Veterans Affairs Foundation for Research and Education, Miami, Florida, United States of America.,Department of Pathology, University of Miami, Miller School of Medicine, Miami, Florida, United States of America.,Department of Medicine, Divisions of Hematology/Oncology and Endocrinology, University of Miami, Miller School of Medicine, Miami, Florida, United States of America.,Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, Florida, United States of America
| | - Lauro M Takeuchi
- Interdisciplinary Stem Cell Institute, University of Miami, Miller School of Medicine, Miami, Florida, United States of America
| | - Petra Popovics
- Veterans Affairs Medical Center and South Florida Veterans Affairs Foundation for Research and Education, Miami, Florida, United States of America.,Department of Medicine III, Medical Faculty Carl Gustav Carus, TU Dresden, Germany.,Department of Medicine, Division of Cardiology, University of Miami, Miller School of Medicine, Miami, Florida, United States of America
| | - Miklos Jaszberenyi
- Veterans Affairs Medical Center and South Florida Veterans Affairs Foundation for Research and Education, Miami, Florida, United States of America.,Department of Pathology, University of Miami, Miller School of Medicine, Miami, Florida, United States of America
| | - Irving Vidaurre
- Veterans Affairs Medical Center and South Florida Veterans Affairs Foundation for Research and Education, Miami, Florida, United States of America
| | - Marta Zarandi
- Veterans Affairs Medical Center and South Florida Veterans Affairs Foundation for Research and Education, Miami, Florida, United States of America
| | - Ren-Zhi Cai
- Veterans Affairs Medical Center and South Florida Veterans Affairs Foundation for Research and Education, Miami, Florida, United States of America.,Department of Pathology, University of Miami, Miller School of Medicine, Miami, Florida, United States of America
| | - Norman L Block
- Interdisciplinary Stem Cell Institute, University of Miami, Miller School of Medicine, Miami, Florida, United States of America.,Veterans Affairs Medical Center and South Florida Veterans Affairs Foundation for Research and Education, Miami, Florida, United States of America.,Department of Pathology, University of Miami, Miller School of Medicine, Miami, Florida, United States of America.,Department of Medicine, Divisions of Hematology/Oncology and Endocrinology, University of Miami, Miller School of Medicine, Miami, Florida, United States of America.,Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, Florida, United States of America
| | - Joshua M Hare
- Interdisciplinary Stem Cell Institute, University of Miami, Miller School of Medicine, Miami, Florida, United States of America.,Department of Medicine, Division of Cardiology, University of Miami, Miller School of Medicine, Miami, Florida, United States of America
| | - Ferenc G Rick
- Veterans Affairs Medical Center and South Florida Veterans Affairs Foundation for Research and Education, Miami, Florida, United States of America.,Department of Urology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida, United States of America
| |
Collapse
|
22
|
McClure C, Ali E, Youssef D, Yao ZQ, McCall CE, El Gazzar M. NFI-A disrupts myeloid cell differentiation and maturation in septic mice. J Leukoc Biol 2015; 99:201-11. [PMID: 26259914 DOI: 10.1189/jlb.4a0415-171rr] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Accepted: 07/27/2015] [Indexed: 12/17/2022] Open
Abstract
Mounting evidence supports that sepsis-associated immunosuppression increases mortality. As potential contributors to poor sepsis outcomes, myeloid-derived suppressor cells, which are Gr1(+) CD11b(+) innate-immune cell progenitors unable to differentiate and possess suppressive activities, expand dramatically in septic mice by a process requiring increased microRNA-21 and microRNA-181b expression. The inhibition of these microRNAs in vivo in septic mice restores Gr1(+) CD11b(+) cell differentiation and maturation and improves survival. Here, we show that during sepsis-induced generation of myeloid-derived suppressor cells, transcription factor nuclear factor 1 A type represses cyclin-dependent kinase inhibitor p21 to arrest differentiation of Gr1(+) CD11b(+) cells. Our findings include the following: 1) Gr1(+) CD11b(+) myeloid cells from late septic mice genetically lacking nuclear factor 1 A type cannot suppress CD4(+) T cell proliferation and activation; 2) the reconstitution of nuclear factor 1 A type in microRNA-21 and microRNA-181b-depleted Gr1(+) CD11b(+) myeloid-derived suppressor cells inhibits cyclin-dependent kinase inhibitor p21 and restores the immune-suppressor phenotype; 3) ex vivo nuclear factor 1 A type knockdown in Gr1(+) CD11b(+) myeloid-derived suppressor cells from late septic mice restores cyclin-dependent kinase inhibitor p21 expression and promotes monocyte and dendritic cell differentiation; and 4) ectopic nuclear factor 1 A type expression in normal Gr1(+) CD11b(+) cells generates an immunosuppressive phenotype. We suggest that therapeutically targeting nuclear factor 1 A type during late sepsis might improve survival.
Collapse
Affiliation(s)
- Clara McClure
- *Departments of Internal Medicine and Center for Inflammation, Infectious Diseases and Immunity, East Tennessee State University College of Medicine, Johnson City, Tennessee, USA; and Department of Internal Medicine, Section of Molecular Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Ekram Ali
- *Departments of Internal Medicine and Center for Inflammation, Infectious Diseases and Immunity, East Tennessee State University College of Medicine, Johnson City, Tennessee, USA; and Department of Internal Medicine, Section of Molecular Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Dima Youssef
- *Departments of Internal Medicine and Center for Inflammation, Infectious Diseases and Immunity, East Tennessee State University College of Medicine, Johnson City, Tennessee, USA; and Department of Internal Medicine, Section of Molecular Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Zhi Q Yao
- *Departments of Internal Medicine and Center for Inflammation, Infectious Diseases and Immunity, East Tennessee State University College of Medicine, Johnson City, Tennessee, USA; and Department of Internal Medicine, Section of Molecular Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Charles E McCall
- *Departments of Internal Medicine and Center for Inflammation, Infectious Diseases and Immunity, East Tennessee State University College of Medicine, Johnson City, Tennessee, USA; and Department of Internal Medicine, Section of Molecular Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Mohamed El Gazzar
- *Departments of Internal Medicine and Center for Inflammation, Infectious Diseases and Immunity, East Tennessee State University College of Medicine, Johnson City, Tennessee, USA; and Department of Internal Medicine, Section of Molecular Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| |
Collapse
|
23
|
Duval C, Zaniolo K, Leclerc S, Salesse C, Guérin SL. Characterization of the human α9 integrin subunit gene: Promoter analysis and transcriptional regulation in ocular cells. Exp Eye Res 2015; 135:146-63. [PMID: 25746835 DOI: 10.1016/j.exer.2015.03.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Revised: 01/26/2015] [Accepted: 03/02/2015] [Indexed: 11/29/2022]
Abstract
α9β1 is the most recent addition to the integrin family of membrane receptors and consequently remains the one that is the least characterized. To better understand how transcription of the human gene encoding the α9 subunit is regulated, we cloned the α9 promoter and characterized the regulatory elements that are required to ensure its transcription. Transfection of α9 promoter/CAT plasmids in primary cultured human corneal epithelial cells (HCECs) and uveal melanoma cell lines demonstrated the presence of both negative and positive regulatory elements along the α9 promoter and positioned the basal α9 promoter to within 118 bp from the α9 mRNA start site. In vitro DNaseI footprinting and in vivo ChIP analyses demonstrated the binding of the transcription factors Sp1, c-Myb and NFI to the most upstream α9 negative regulatory element. The transcription factors Sp1 and NFI were found to bind the basal α9 promoter individually but Sp1 binding clearly predominates when both transcription factors are present in the same extract. Suppression of Sp1 expression through RNAi also caused a dramatic reduction in the expression of the α9 gene. Most of all, addition of tenascin-C (TNC), the ligand of α9β1, to the tissue culture plates prior to seeding HCECs increased α9 transcription whereas it simultaneously decreased expression of the α5 integrin subunit gene. This dual regulatory action of TNC on the transcription of the α9 and α5 genes suggests that both these integrins must work together to appropriately regulate cell adhesion, migration and differentiation that are hallmarks of tissue wound healing.
Collapse
Affiliation(s)
- Céline Duval
- Centre Universitaire d'Ophtalmologie-Recherche, Axe Médecine Régénératrice, Hôpital du Saint-Sacrement, Centre de Recherche FRQS du CHU de Québec, Québec, Canada
| | - Karine Zaniolo
- Centre Universitaire d'Ophtalmologie-Recherche, Axe Médecine Régénératrice, Hôpital du Saint-Sacrement, Centre de Recherche FRQS du CHU de Québec, Québec, Canada
| | - Steeve Leclerc
- Centre Universitaire d'Ophtalmologie-Recherche, Axe Médecine Régénératrice, Hôpital du Saint-Sacrement, Centre de Recherche FRQS du CHU de Québec, Québec, Canada
| | - Christian Salesse
- Centre Universitaire d'Ophtalmologie-Recherche, Axe Médecine Régénératrice, Hôpital du Saint-Sacrement, Centre de Recherche FRQS du CHU de Québec, Québec, Canada; Département d'Ophtalmologie, Faculté de Médecine, Université Laval, Québec, QC, Canada
| | - Sylvain L Guérin
- Centre Universitaire d'Ophtalmologie-Recherche, Axe Médecine Régénératrice, Hôpital du Saint-Sacrement, Centre de Recherche FRQS du CHU de Québec, Québec, Canada; Département d'Ophtalmologie, Faculté de Médecine, Université Laval, Québec, QC, Canada.
| |
Collapse
|
24
|
Chen Q, Lu H, Yang H. Chitosan inhibits fibroblasts growth in Achilles tendon via TGF-β1/Smad3 pathway by miR-29b. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2014; 7:8462-8470. [PMID: 25674210 PMCID: PMC4313955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 12/01/2014] [Indexed: 06/04/2023]
Abstract
BACKGROUND AND AIM Chitosan, is a natural polymer, plays an important role in prevention of tendon adhesion in tendon healing process. However, the molecular mechanisms underlying the prevention effect is unclear. Here we investigated the effects of chitosan on Achilles tendon injury rats and fibroblasts. METHODS Eight weeks after surgery, gliding excursion and the content of collagen fibers in Achilles tendon injury rats were determined to evaluate the chitosan effect on tendon healing. Fibroblasts isolated from scar tissue of repaired tendon were treated with different concentration of chitosan, and then cell inhibition, apoptosis and cell cycle were measured using MTT and Flow Cytometry respectively. The expression of microRNAs (miRNAs) was quantified by real-time PCR and protein expression of TGF-β1, Smad3 and P21 were quantified by western blotting. MiR-29b inhibitor was transfected in cells to evaluate the mechanism underlying the effects of chitosan on tendon fibroblasts. RESULTS The gliding excursion of repaired tendon was increased and the content of collagen fibers was decreased by chitosan in rats. Chitosan inhibited the fibroblasts growth and arrested cells in G1 phase. Chitosan also elevated the expression of miR-29b and P21 while reduced the levels of TGFβ1 and Smad3 in both repaired tendon and fibroblasts. In addition, miR-29b inhibitor revered the effects of chitosan on fibroblasts. CONCLUSIONS The current study demonstrated that chitosan improving the condition of tendon healing after surgery, which is reduced by the high expression of miR-29b and its down-regulation of TGF-β1/Smad3 level and inhibition of fibroblasts growth.
Collapse
Affiliation(s)
- Qiang Chen
- Department of Hand Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University China
| | - Hui Lu
- Department of Hand Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University China
| | - Hu Yang
- Department of Hand Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University China
| |
Collapse
|
25
|
Bonacini M, Coletta M, Ramazzina I, Naponelli V, Modernelli A, Davalli P, Bettuzzi S, Rizzi F. Distinct promoters, subjected to epigenetic regulation, drive the expression of two clusterin mRNAs in prostate cancer cells. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2014; 1849:44-54. [PMID: 25464035 DOI: 10.1016/j.bbagrm.2014.11.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Revised: 10/24/2014] [Accepted: 11/03/2014] [Indexed: 01/13/2023]
Abstract
The human clusterin (CLU) gene codes for several mRNAs characterized by different sequences at their 5' end. We investigated the expression of two CLU mRNAs, called CLU 1 and CLU 2, in immortalized (PNT1a) and tumorigenic (PC3 and DU145) prostate epithelial cells, as well as in normal fetal fibroblasts (WI38) following the administration of the epigenetic drugs 5-aza-2'-deoxycytidine (AZDC) and trichostatin A (TSA) given either as single or combined treatment (AZDC-TSA). Our experimental evidences show that: a) CLU 1 is the most abundant transcript variant. b) CLU 2 is expressed at a low level in normal fibroblasts and virtually absent in prostate cancer cells. c) CLU 1, and to a greater extent CLU 2 expression, increased by AZDC-TSA treatment in prostate cancer cells. d) Both CLU 1 and CLU 2 encode for secreted CLU. e) P2, a novel promoter that overlaps the CLU 2 Transcription Start Site (TSS), drives CLU 2 expression. f) A CpG island, methylated in prostate cancer cells and not in normal fibroblasts, is responsible for long-term heritable regulation of CLU 1 expression. g) ChIP assay of histone tail modifications at CLU promoters (P1 and P2) shows that treatment of prostate cancer cells with AZDC-TSA causes enrichment of Histone3(Lys9)acetylated (H3K9ac) and reduction of Histone3(Lys27)trimethylated (H3K27me3), inducing active transcription of both CLU variants. In conclusion, we show for the first time that the expression of CLU 2 mRNA is driven by a novel promoter, P2, whose activity responds to epigenetic drugs treatment through changes in histone modifications.
Collapse
Affiliation(s)
- Martina Bonacini
- Department of Biomedicine, Biotechnology and Translational Research, University of Parma, Via Volturno 39/a, 43126 Parma, Italy
| | - Mariangela Coletta
- Department of Biomedicine, Biotechnology and Translational Research, University of Parma, Via Volturno 39/a, 43126 Parma, Italy
| | - Ileana Ramazzina
- Department of Biomedicine, Biotechnology and Translational Research, University of Parma, Via Volturno 39/a, 43126 Parma, Italy; Centre for Molecular and Translational Oncology (COMT), University of Parma, Parco Area delle Scienze 11/a, 43124 Parma, Italy; National Institute of Biostructure and Biosystems (INBB), Viale Medaglie d'Oro 305, 00136 Rome, Italy
| | - Valeria Naponelli
- Department of Biomedicine, Biotechnology and Translational Research, University of Parma, Via Volturno 39/a, 43126 Parma, Italy; Centre for Molecular and Translational Oncology (COMT), University of Parma, Parco Area delle Scienze 11/a, 43124 Parma, Italy; National Institute of Biostructure and Biosystems (INBB), Viale Medaglie d'Oro 305, 00136 Rome, Italy
| | - Alice Modernelli
- Department of Biomedicine, Biotechnology and Translational Research, University of Parma, Via Volturno 39/a, 43126 Parma, Italy
| | - Pierpaola Davalli
- Department of Biomedical Sciences, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via Campi 287, 41125 Modena, Italy
| | - Saverio Bettuzzi
- Department of Biomedicine, Biotechnology and Translational Research, University of Parma, Via Volturno 39/a, 43126 Parma, Italy; Centre for Molecular and Translational Oncology (COMT), University of Parma, Parco Area delle Scienze 11/a, 43124 Parma, Italy; National Institute of Biostructure and Biosystems (INBB), Viale Medaglie d'Oro 305, 00136 Rome, Italy.
| | - Federica Rizzi
- Department of Biomedicine, Biotechnology and Translational Research, University of Parma, Via Volturno 39/a, 43126 Parma, Italy; Centre for Molecular and Translational Oncology (COMT), University of Parma, Parco Area delle Scienze 11/a, 43124 Parma, Italy; National Institute of Biostructure and Biosystems (INBB), Viale Medaglie d'Oro 305, 00136 Rome, Italy
| |
Collapse
|
26
|
Deák F, Mátés L, Korpos E, Zvara A, Szénási T, Kiricsi M, Mendler L, Keller-Pintér A, Ozsvári B, Juhász H, Sorokin L, Dux L, Mermod N, Puskás LG, Kiss I. Extracellular deposition of matrilin-2 controls the timing of the myogenic program during muscle regeneration. J Cell Sci 2014; 127:3240-56. [PMID: 24895400 PMCID: PMC4117230 DOI: 10.1242/jcs.141556] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2013] [Accepted: 05/08/2014] [Indexed: 01/05/2023] Open
Abstract
Here, we identify a role for the matrilin-2 (Matn2) extracellular matrix protein in controlling the early stages of myogenic differentiation. We observed Matn2 deposition around proliferating, differentiating and fusing myoblasts in culture and during muscle regeneration in vivo. Silencing of Matn2 delayed the expression of the Cdk inhibitor p21 and of the myogenic genes Nfix, MyoD and Myog, explaining the retarded cell cycle exit and myoblast differentiation. Rescue of Matn2 expression restored differentiation and the expression of p21 and of the myogenic genes. TGF-β1 inhibited myogenic differentiation at least in part by repressing Matn2 expression, which inhibited the onset of a positive-feedback loop whereby Matn2 and Nfix activate the expression of one another and activate myoblast differentiation. In vivo, myoblast cell cycle arrest and muscle regeneration was delayed in Matn2(-/-) relative to wild-type mice. The expression levels of Trf3 and myogenic genes were robustly reduced in Matn2(-/-) fetal limbs and in differentiating primary myoblast cultures, establishing Matn2 as a key modulator of the regulatory cascade that initiates terminal myogenic differentiation. Our data thus identify Matn2 as a crucial component of a genetic switch that modulates the onset of tissue repair.
Collapse
Affiliation(s)
- Ferenc Deák
- Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, Szeged, H-6701 Szeged, Hungary
| | - Lajos Mátés
- Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, Szeged, H-6701 Szeged, Hungary Institute of Genetics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, H-6701 Szeged, Hungary
| | - Eva Korpos
- Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, Szeged, H-6701 Szeged, Hungary Institute of Physiological Chemistry and Pathobiochemistry, Muenster University, D-48149 Muenster, Germany
| | - Agnes Zvara
- Institute of Genetics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, H-6701 Szeged, Hungary
| | - Tibor Szénási
- Institute of Genetics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, H-6701 Szeged, Hungary
| | - Mónika Kiricsi
- Institute of Biochemistry, Faculty of General Medicine, University of Szeged, H-6720 Szeged, Hungary Department of Biochemistry and Molecular Biology, Faculty of Natural Sciences and Informatics, University of Szeged, H-6720 Szeged, Hungary
| | - Luca Mendler
- Institute of Biochemistry, Faculty of General Medicine, University of Szeged, H-6720 Szeged, Hungary
| | - Anikó Keller-Pintér
- Institute of Biochemistry, Faculty of General Medicine, University of Szeged, H-6720 Szeged, Hungary
| | | | - Hajnalka Juhász
- Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, Szeged, H-6701 Szeged, Hungary
| | - Lydia Sorokin
- Institute of Physiological Chemistry and Pathobiochemistry, Muenster University, D-48149 Muenster, Germany
| | - László Dux
- Institute of Biochemistry, Faculty of General Medicine, University of Szeged, H-6720 Szeged, Hungary
| | - Nicolas Mermod
- Institute of Biotechnology, University of Lausanne, and Center for Biotechnology of the University of Lausanne and École Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | - László G Puskás
- Institute of Genetics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, H-6701 Szeged, Hungary Avidin Ltd., H-6726 Szeged, Hungary
| | - Ibolya Kiss
- Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, Szeged, H-6701 Szeged, Hungary Avidin Ltd., H-6726 Szeged, Hungary
| |
Collapse
|
27
|
MicroRNA 21 (miR-21) and miR-181b couple with NFI-A to generate myeloid-derived suppressor cells and promote immunosuppression in late sepsis. Infect Immun 2014; 82:3816-25. [PMID: 24980967 DOI: 10.1128/iai.01495-14] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
The sepsis initial hyperinflammatory reaction, if not treated early, shifts to a protracted state of immunosuppression that alters both innate and adaptive immunity and is associated with elevated mortality. Myeloid-derived suppressor cells (MDSCs) are myeloid progenitors and precursors that fail to differentiate into mature innate-immunity cells and are known for their potent immunosuppressive activities. We previously reported that murine MDSCs expand dramatically in the bone marrow during late sepsis, induced by cecal ligation and puncture, and demonstrated that they contribute to late-sepsis immunosuppression. However, the molecular mechanism responsible for generating these immature Gr1(+) CD11b(+) myeloid cells during sepsis remains unknown. We show here that sepsis generates a microRNA (miRNA) signature that expands MDSCs. We found that miRNA 21 (miR-21) and miR-181b expression is upregulated in early sepsis and sustained in late sepsis. Importantly, we found that simultaneous in vivo blockade of both miRNAs via antagomiR (a chemically modified miRNA inhibitor) injection after sepsis initiation decreased the bone marrow Gr1(+) CD11b(+) myeloid progenitors, improved bacterial clearance, and reduced late-sepsis mortality by 74%. Gr1(+) CD11b(+) cells isolated from mice injected with antagomiRs were able to differentiate ex vivo into macrophages and dendritic cells and produced smaller amounts of the immunosuppressive interleukin 10 (IL-10) and transforming growth factor β (TGF-β) after stimulation with bacterial lipopolysaccharide, suggesting that immature myeloid cells regained their maturation potential and have lost their immunosuppressive activity. In addition, we found that the protein level of transcription factor NFI-A, which plays a role in myeloid cell differentiation, was increased during sepsis and that antagomiR injection reduced its expression. Moreover, knockdown of NFI-A in the Gr1(+) CD11b(+) cells isolated from late-septic mice increased their maturation potential and reduced their production of the immunosuppressive mediators, similar to antagomiR injection. These data support the hypothesis that sepsis reprograms myeloid cells and thus alters the innate immunity cell repertoire to promote immunosuppression, and they demonstrate that this process can be reversed by targeting miR-21 and miR-181b to improve late-sepsis survival.
Collapse
|
28
|
Lee JS, Xiao J, Patel P, Schade J, Wang J, Deneen B, Erdreich-Epstein A, Song HR. A novel tumor-promoting role for nuclear factor IA in glioblastomas is mediated through negative regulation of p53, p21, and PAI1. Neuro Oncol 2013; 16:191-203. [PMID: 24305710 DOI: 10.1093/neuonc/not167] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Background Nuclear factor IA (NFIA), a transcription factor and essential regulator in embryonic glial development, is highly expressed in human glioblastoma (GBM) compared with normal brain, but its contribution to GBM and cancer pathogenesis is unknown. Here we demonstrate a novel role for NFIA in promoting growth and migration of GBM and establish the molecular mechanisms mediating these functions. Methods To determine the role of NFIA in glioma, we examined the effects of NFIA in growth, proliferation, apoptosis, and migration. We used gain-of-function (overexpression) and loss-of-function (shRNA knockdown) of NFIA in primary patient-derived GBM cells and established glioma cell lines in culture and in intracranial xenografts in mouse brains. Results Knockdown of native NFIA blocked tumor growth and induced cell death and apoptosis. Complementing this, NFIA overexpression accelerated growth, proliferation, and migration of GBM in cell culture and in mouse brains. These NFIA tumor-promoting effects were mediated via transcriptional repression of p53, p21, and plasminogen activator inhibitor 1 (PAI1) through specific NFIA-recognition sequences in their promoters. Importantly, the effects of NFIA on proliferation and apoptosis were independent of TP53 mutation status, a finding especially relevant for GBM, in which TP53 is frequently mutated. Conclusion NFIA is a modulator of GBM growth and migration, and functions by distinct regulation of critical oncogenic pathways that govern the malignant behavior of GBM.
Collapse
Affiliation(s)
- Jun Sung Lee
- Corresponding author: Hae-Ri Song, MD, New York University School of Medicine, Smilow Research Center 1306, 522 First Avenue, New York, NY 10016.
| | | | | | | | | | | | | | | |
Collapse
|
29
|
The miR-223/nuclear factor I-A axis regulates glial precursor proliferation and tumorigenesis in the CNS. J Neurosci 2013; 33:13560-8. [PMID: 23946414 DOI: 10.1523/jneurosci.0321-13.2013] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Contemporary views of tumorigenesis regard its inception as a convergence of genetic mutation and developmental context. Glioma is the most common and deadly malignancy in the CNS; therefore, understanding how regulators of glial development contribute to its formation remains a key question. Previously we identified nuclear factor I-A (NFIA) as a key regulator of developmental gliogenesis, while miR-223 has been shown to repress NFIA expression in other systems. Using this relationship as a starting point, we found that miR-223 can suppress glial precursor proliferation via repression of NFIA during chick spinal cord development. This relationship is conserved in glioma, as miR-223 and NFIA expression is negatively correlated in human glioma tumors, and the miR-223/NFIA axis suppresses tumorigenesis in a human glioma cell line. Subsequent analysis of NFIA function revealed that it directly represses p21 and is required for tumorigenesis in a mouse neural stem cell model of glioma. These studies represent the first characterization of miR-223/NFIA axis function in glioma and demonstrate that it is a conserved proliferative mechanism across CNS development and tumorigenesis.
Collapse
|
30
|
Dellago H, Preschitz‐Kammerhofer B, Terlecki‐Zaniewicz L, Schreiner C, Fortschegger K, Chang MW, Hackl M, Monteforte R, Kühnel H, Schosserer M, Gruber F, Tschachler E, Scheideler M, Grillari‐Voglauer R, Grillari J, Wieser M. High levels of oncomiR-21 contribute to the senescence-induced growth arrest in normal human cells and its knock-down increases the replicative lifespan. Aging Cell 2013; 12:446-58. [PMID: 23496142 PMCID: PMC3864473 DOI: 10.1111/acel.12069] [Citation(s) in RCA: 88] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/21/2013] [Indexed: 12/19/2022] Open
Abstract
Cellular senescence of normal human cells has by now far exceeded its initial role as a model system for aging research. Many reports show the accumulation of senescent cells in vivo, their effect on their microenvironment and its double-edged role as tumour suppressor and promoter. Importantly, removal of senescent cells delays the onset of age-associated diseases in mouse model systems. To characterize the role of miRNAs in cellular senescence of endothelial cells, we performed miRNA arrays from HUVECs of five different donors. Twelve miRNAs, comprising hsa-miR-23a, hsa-miR-23b, hsa-miR-24, hsa-miR-27a, hsa-miR-29a, hsa-miR-31, hsa-miR-100, hsa-miR-193a, hsa-miR-221, hsa-miR-222 and hsa-let-7i are consistently up-regulated in replicatively senescent cells. Surprisingly, also miR-21 was found up-regulated by replicative and stress-induced senescence, despite being described as oncogenic. Transfection of early passage endothelial cells with miR-21 resulted in lower angiogenesis, and less cell proliferation mirrored by up-regulation of p21CIP1 and down-regulation of CDK2. These two cell-cycle regulators are indirectly regulated by miR-21 via its validated direct targets NFIB (Nuclear factor 1 B-type), a transcriptional inhibitor of p21CIP1, and CDC25A, which regulates CDK2 activity by dephosphorylation. Knock-down of either NFIB or CDC25A shows a phenocopy of over-expressing miR-21 in regard to cell-cycle arrest. Finally, miR-21 over-epxression reduces the replicative lifespan, while stable knock-down by sponges extends the replicative lifespan of endothelial cells. Therefore, we propose that miR-21 is the first miRNA that upon its knock-down extends the replicative lifespan of normal human cells.
Collapse
Affiliation(s)
- Hanna Dellago
- Department of Biotechnology BOKU‐VIBT University of Natural Resources and Life Sciences Vienna Muthgasse 181190Vienna Austria
| | - Barbara Preschitz‐Kammerhofer
- Department of Biotechnology BOKU‐VIBT University of Natural Resources and Life Sciences Vienna Muthgasse 181190Vienna Austria
| | - Lucia Terlecki‐Zaniewicz
- Department of Biotechnology BOKU‐VIBT University of Natural Resources and Life Sciences Vienna Muthgasse 181190Vienna Austria
| | - Carina Schreiner
- Department of Biotechnology BOKU‐VIBT University of Natural Resources and Life Sciences Vienna Muthgasse 181190Vienna Austria
| | - Klaus Fortschegger
- CCRI ‐ Children's Cancer Research Institute Zimmermannplatz 101090 Vienna Austria
| | - Martina W.‐F. Chang
- Department of Biotechnology BOKU‐VIBT University of Natural Resources and Life Sciences Vienna Muthgasse 181190Vienna Austria
| | - Matthias Hackl
- Department of Biotechnology BOKU‐VIBT University of Natural Resources and Life Sciences Vienna Muthgasse 181190Vienna Austria
| | - Rossella Monteforte
- Department of Biotechnology BOKU‐VIBT University of Natural Resources and Life Sciences Vienna Muthgasse 181190Vienna Austria
| | - Harald Kühnel
- Institute of Physiology, Pathophysiology and Biophysics Department of Biomedical Sciences University of Veterinary Medicine Vienna Veterinärplatz 1A‐1210Vienna Austria
| | - Markus Schosserer
- Department of Biotechnology BOKU‐VIBT University of Natural Resources and Life Sciences Vienna Muthgasse 181190Vienna Austria
| | - Florian Gruber
- Department of Dermatology Medical University of Vienna A‐1090Vienna Austria
| | - Erwin Tschachler
- Department of Dermatology Medical University of Vienna A‐1090Vienna Austria
- C.E.R.I.E.S. ‐ Centre de Recherches et d'Investigations Epidermiques et Sensorielles 20 Rue Victor Noir 92200 Neuilly‐sur‐Seine France
| | - Marcel Scheideler
- Institute for Genomics and Bioinformatics Graz University of Technology Petersgasse 148010Graz Austria
| | - Regina Grillari‐Voglauer
- Department of Biotechnology BOKU‐VIBT University of Natural Resources and Life Sciences Vienna Muthgasse 181190Vienna Austria
- Evercyte GmbH Muthgasse 181190Vienna Austria
- Austrian Centre of Industrial Biotechnology (ACIB GmbH) Muthgasse 181190Vienna Austria
| | - Johannes Grillari
- Department of Biotechnology BOKU‐VIBT University of Natural Resources and Life Sciences Vienna Muthgasse 181190Vienna Austria
- Evercyte GmbH Muthgasse 181190Vienna Austria
| | - Matthias Wieser
- Department of Biotechnology BOKU‐VIBT University of Natural Resources and Life Sciences Vienna Muthgasse 181190Vienna Austria
- Austrian Centre of Industrial Biotechnology (ACIB GmbH) Muthgasse 181190Vienna Austria
| |
Collapse
|
31
|
Gato WE, Hales DB, Means JC. Hepatic gene expression analysis of 2-aminoanthracene exposed Fisher-344 rats reveal patterns indicative of liver carcinoma and type 2 diabetes. J Toxicol Sci 2013; 37:1001-16. [PMID: 23038007 DOI: 10.2131/jts.37.1001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
The goal of the present study was to examine hepatic differential gene expression patterns in Fisher-344 rats in response to dietary 2-aminoanthracene (2AA) ingestion for 14 and 28 days. Twenty four post-weaning 3-4 week old F-344 male rats were exposed to 0 mgkg(-1)-diet (control), 50 mgkg(-1)-diet (low dose), 75 mgkg(-1)-diet (medium dose) and 100 mgkg(-1)-diet (high dose) 2AA for 14 and 28 days. This was followed by analysis of the liver for global gene expression changes. In both time points, the numbers of genes affected seem to correlate with the dose of 2AA. Sixteen mRNAs were differentially expressed in all treatment groups for the short-term exposure group. Similarly, 51 genes were commonly expressed in all 28-day exposure group. Almost all the genes seem to have higher expression relative to the controls. In contrast, cytochrome P450 family 4, subfamily a, polypeptide 8 (Cyp4a8), and monocyte to macrophage differentiation-associated (Mmd2) were down-regulated relative to controls. Differentially expressed mRNAs were further analyzed for associations via DAVID. GO categories show the effect of 2AA to be linked with genes responsible for carbohydrate utilization and transport, lipid metabolic processes, stress responses such as inflammation and apoptosis processes, immune system response, DNA damage response, cancer processes and circadian rhythm. The data from the current study identified altered hepatic gene expression profiles that may be associated with carcinoma, autoimmune response, and/or type 2 diabetes. Possible biomarkers due to 2AA toxicity in the liver for future study include Abcb1a, Nhej1, Adam8, Cdkn1a, Mgmt, and Nrcam.
Collapse
Affiliation(s)
- Worlanyo E Gato
- Department of Chemistry and Biochemistry, Southern Illinois University Carbondale, IL 62901, USA
| | | | | |
Collapse
|
32
|
Pjanic M, Schmid CD, Gaussin A, Ambrosini G, Adamcik J, Pjanic P, Plasari G, Kerschgens J, Dietler G, Bucher P, Mermod N. Nuclear Factor I genomic binding associates with chromatin boundaries. BMC Genomics 2013; 14:99. [PMID: 23402308 PMCID: PMC3610271 DOI: 10.1186/1471-2164-14-99] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2012] [Accepted: 01/26/2013] [Indexed: 12/20/2022] Open
Abstract
Background The Nuclear Factor I (NFI) family of DNA binding proteins (also called CCAAT box transcription factors or CTF) is involved in both DNA replication and gene expression regulation. Using chromatin immuno-precipitation and high throughput sequencing (ChIP-Seq), we performed a genome-wide mapping of NFI DNA binding sites in primary mouse embryonic fibroblasts. Results We found that in vivo and in vitro NFI DNA binding specificities are indistinguishable, as in vivo ChIP-Seq NFI binding sites matched predictions based on previously established position weight matrix models of its in vitro binding specificity. Combining ChIP-Seq with mRNA profiling data, we found that NFI preferentially associates with highly expressed genes that it up-regulates, while binding sites were under-represented at expressed but unregulated genes. Genomic binding also correlated with markers of transcribed genes such as histone modifications H3K4me3 and H3K36me3, even outside of annotated transcribed loci, implying NFI in the control of the deposition of these modifications. Positional correlation between + and - strand ChIP-Seq tags revealed that, in contrast to other transcription factors, NFI associates with a nucleosomal length of cleavage-resistant DNA, suggesting an interaction with positioned nucleosomes. In addition, NFI binding prominently occurred at boundaries displaying discontinuities in histone modifications specific of expressed and silent chromatin, such as loci submitted to parental allele-specific imprinted expression. Conclusions Our data thus suggest that NFI nucleosomal interaction may contribute to the partitioning of distinct chromatin domains and to epigenetic gene expression regulation. NFI ChIP-Seq and input control DNA data were deposited at Gene Expression Omnibus (GEO) repository under accession number GSE15844. Gene expression microarray data for mouse embryonic fibroblasts are on GEO accession number GSE15871.
Collapse
Affiliation(s)
- Milos Pjanic
- Institute of Biotechnology and Center for Biotecghnology UNIL-EPFL, University of Lausanne, 1015, Lausanne, Switzerland.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Ross RW, Galsky MD, Scher HI, Magidson J, Wassmann K, Lee GSM, Katz L, Subudhi SK, Anand A, Fleisher M, Kantoff PW, Oh WK. A whole-blood RNA transcript-based prognostic model in men with castration-resistant prostate cancer: a prospective study. Lancet Oncol 2012; 13:1105-13. [PMID: 23059047 DOI: 10.1016/s1470-2045(12)70263-2] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Survival for patients with castration-resistant prostate cancer is highly variable. We assessed the effectiveness of a whole-blood RNA transcript-based model as a prognostic biomarker in castration-resistant prostate cancer. METHODS Peripheral blood was prospectively collected from 62 men with castration-resistant prostate cancer on various treatment regimens who were enrolled in a training set at the Dana-Farber Cancer Institute (Boston, MA, USA) from August, 2006, to June, 2008, and from 140 patients with castration-resistant prostate cancer in a validation set from Memorial Sloan-Kettering Cancer Center (New York, NY, USA) from August, 2006, to February, 2009. A panel of 168 inflammation-related and prostate cancer-related genes was assessed with optimised quantitative PCR to assess biomarkers predictive of survival. FINDINGS A six-gene model (consisting of ABL2, SEMA4D, ITGAL, and C1QA, TIMP1, CDKN1A) separated patients with castration-resistant prostate cancer into two risk groups: a low-risk group with a median survival of more than 34·9 months (median survival was not reached) and a high-risk group with a median survival of 7·8 months (95% CI 1·8-13·9; p<0·0001). The prognostic utility of the six-gene model was validated in an independent cohort. This model was associated with a significantly higher area under the curve compared with a clinicopathological model (0·90 [95% CI 0·78-0·96] vs 0·65 [0·52-0·78]; p=0·0067). INTERPRETATION Transcriptional profiling of whole blood yields crucial prognostic information about men with castration-resistant prostate cancer. The six-gene model suggests possible dysregulation of the immune system, a finding that warrants further study. FUNDING Source MDX.
Collapse
Affiliation(s)
- Robert W Ross
- Division of Solid Tumor Oncology, Department of Medicine, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Duval C, Gaudreault M, Vigneault F, Touzel-Deschênes L, Rochette PJ, Masson-Gadais B, Germain L, Guérin SL. Rescue of the transcription factors Sp1 and NFI in human skin keratinocytes through a feeder-layer-dependent suppression of the proteasome activity. J Mol Biol 2012; 418:281-99. [PMID: 22420942 DOI: 10.1016/j.jmb.2012.01.021] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2011] [Revised: 01/17/2012] [Accepted: 01/19/2012] [Indexed: 12/01/2022]
Abstract
Co-culturing human skin keratinocytes along with a feeder layer has proven to considerably improve their proliferative properties by delaying massive induction of terminal differentiation. Through a yet unclear mechanism, we recently reported that irradiated 3T3 (i3T3) fibroblasts used as a feeder layer increase the nuclear content of Sp1, a positive transcription factor (TF) that plays a critical role in many cellular functions including cell proliferation, into both adult skin keratinocytes and newborn skin keratinocytes. In this study, we examined the influence of i3T3 on the expression and DNA binding of NFI, another TF important for cell proliferation and cell cycle progression, and attempted to decipher the mechanism by which the feeder layer contributes at maintaining higher levels of these TFs in skin keratinocytes. Our results indicate that co-culturing both adult skin keratinocytes and newborn skin keratinocytes along with a feeder layer dramatically increases glycosylation of NFI and may prevent it from being degraded by the proteasome.
Collapse
Affiliation(s)
- Céline Duval
- LOEX/CUO-Recherche, Hôpital du Saint-Sacrement, Centre de Recherche du CHA, Québec, QC, Canada G1S4L8
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Lee DS, Yoon WJ, Cho ES, Kim HJ, Gronostajski RM, Cho MI, Park JC. Crosstalk between nuclear factor I-C and transforming growth factor-β1 signaling regulates odontoblast differentiation and homeostasis. PLoS One 2011; 6:e29160. [PMID: 22195013 PMCID: PMC3241690 DOI: 10.1371/journal.pone.0029160] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2011] [Accepted: 11/22/2011] [Indexed: 02/02/2023] Open
Abstract
Transforming growth factor-β1 (TGF-β1) signaling plays a key role in vertebrate development, homeostasis, and disease. Nuclear factor I-C (NFI-C) has been implicated in TGF-β1 signaling, extracellular matrix gene transcription, and tooth root development. However, the functional relationship between NFI-C and TGF-β1 signaling remains uncharacterized. The purpose of this study was to identify the molecular interactions between NFI-C and TGF-β1 signaling in mouse odontoblasts. Real-time polymerase chain reaction and western analysis demonstrated that NFI-C expression levels were inversely proportional to levels of TGF-β1 signaling molecules during in vitro odontoblast differentiation. Western blot and immunofluorescence results showed that NFI-C was significantly degraded after TGF-β1 addition in odontoblasts, and the formation of the Smad3 complex was essential for NFI-C degradation. Additionally, ubiquitination assay results showed that Smurf1 and Smurf2 induced NFI-C degradation and polyubiquitination in a TGF-β1-dependent manner. Both kinase and in vitro binding assays revealed that the interaction between NFI-C and Smurf1/Smurf2 requires the activation of the mitogen-activated protein kinase pathway by TGF-β1. Moreover, degradation of NFI-C induced by TGF-β1 occurred generally in cell types other than odontoblasts in normal human breast epithelial cells. In contrast, NFI-C induced dephosphorylation of p-Smad2/3. These results show that crosstalk between NFI-C and TGF-β1 signaling regulates cell differentiation and homeostatic processes in odontoblasts, which might constitute a common cellular mechanism.
Collapse
Affiliation(s)
- Dong-Seol Lee
- Department of Oral Histology-Developmental Biology and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, Korea
| | - Won-Joon Yoon
- Department of Molecular Genetics, BK21 Project, School of Dentistry, Seoul National University, Seoul, Korea
| | - Eui Sic Cho
- Laboratory for Craniofacial Biology, Institute of Oral Bioscience, Chonbuk National University, Jeon-ju, Korea
| | - Heung-Joong Kim
- Department of Oral Anatomy and Developmental Biology, College of Dentistry, Chosun University, Gwang-ju, Korea
| | - Richard M. Gronostajski
- Department of Biochemistry, Program in Neuroscience, Developmental Genomics Group and Center of Excellence in Bioinformatics and Life Sciences, School of Medicine and Biomedical Science, State University of New York at Buffalo, Buffalo, New York, United States of America
| | - Moon-Il Cho
- Department of Oral Biology, School of Dental Medicine, State University of New York at Buffalo, Buffalo, New York, United States of America
| | - Joo-Cheol Park
- Department of Oral Histology-Developmental Biology and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, Korea
- * E-mail:
| |
Collapse
|
36
|
Choi J, Ababon MR, Matteson PG, Millonig JH. Cut-like homeobox 1 and nuclear factor I/B mediate ENGRAILED2 autism spectrum disorder-associated haplotype function. Hum Mol Genet 2011; 21:1566-80. [PMID: 22180456 DOI: 10.1093/hmg/ddr594] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Both common and rare variants contribute to autism spectrum disorder (ASD) risk, but few variants have been established as functional. Previously we demonstrated that an intronic haplotype (rs1861972-rs1861973 A-C) in the homeobox transcription factor ENGRAILED2 (EN2) is significantly associated with ASD. Positive association has also been reported in six additional data sets, suggesting EN2 is an ASD susceptibility gene. Additional support for this possibility requires identification of functional variants that affect EN2 regulation or activity. In this study, we demonstrate that the A-C haplotype is a transcriptional activator. Luciferase (luc) assays in mouse neuronal cultures determined that the A-C haplotype increases expression levels (50%, P < 0.01, 24 h; 250%, P < 0.0001, 72 h). Mutational analysis indicates that the A-C haplotype activator function requires both associated A and C alleles. A minimal 202-bp element is sufficient for function and also specifically binds a protein complex. Mass spectrometry identified these proteins as the transcription factors, Cut-like homeobox 1 (Cux1) and nuclear factor I/B (Nfib). Subsequent antibody supershifts and chromatin immunoprecipitations demonstrated that human CUX1 and NFIB bind the A-C haplotype. Co-transfection and knock-down experiments determined that both CUX1 and NFIB are required for the A-C haplotype activator function. These data demonstrate that the ASD-associated A-C haplotype is a transcriptional activator, and both CUX1 and NFIB mediate this activity. These results provide biochemical evidence that the ASD-associated A-C haplotype is functional, further supporting EN2 as an ASD susceptibility gene.
Collapse
Affiliation(s)
- Jiyeon Choi
- Center for Advanced Biotechnology and Medicine, Piscataway, NJ 08854, USA
| | | | | | | |
Collapse
|
37
|
Luciakova K, Kollarovic G, Kretova M, Sabova L, Nelson BD. TGF-β signals the formation of a unique NF1/Smad4-dependent transcription repressor-complex in human diploid fibroblasts. Biochem Biophys Res Commun 2011; 411:648-53. [PMID: 21782795 DOI: 10.1016/j.bbrc.2011.07.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2011] [Accepted: 07/03/2011] [Indexed: 10/18/2022]
Abstract
We earlier reported the formation of a unique nuclear NF1/Smad complex in serum-restricted fibroblasts that acts as an NF1-dependent repressor of the human adenine nucleotide translocase-2 gene (ANT2) [K. Luciakova, G. Kollarovic, P. Barath, B.D. Nelson, Growth-dependent repression of human adenine nucleotide translocator-2 (ANT2) transcription: evidence for the participation of Smad and Sp family proteins in the NF1-dependent repressor complex, Biochem. J. 412 (2008) 123-130]. In the present study, we show that TGF-β, like serum-restriction: (a) induces the formation of NF1/Smad repressor complexes, (b) increases binding of the complexes to the repressor elements (Go elements) in the ANT2 promoter, and (c) inhibits ANT2 expression. Repression of ANT2 by TGF-β is eliminated by mutating the NF1 binding sites in the Go repressor elements. All of the above responses to TGF-β are prevented by inhibitors of TGF-β RI and MAPK p38. These inhibitors also prevent NF1/Smad4 repressor complex formation and repression of ANT2 expression in serum-restricted cells, suggesting that similar signaling pathways are initiated by TGF-β and serum-restriction. The present finding that NF1/Smad4 repressor complexes are formed through TGF-β signaling pathways suggests a new, but much broader, role for these complexes in the initiation or maintenance of the growth-inhibited state.
Collapse
Affiliation(s)
- Katarina Luciakova
- Cancer Research Institute, Slovak Academy of Sciences, Bratislava, Vlarska 7, 833 91 Bratislava, Slovak Republic.
| | | | | | | | | |
Collapse
|
38
|
Singh U, Roswall P, Uhrbom L, Westermark B. CGGBP1 regulates cell cycle in cancer cells. BMC Mol Biol 2011; 12:28. [PMID: 21733196 PMCID: PMC3142506 DOI: 10.1186/1471-2199-12-28] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2011] [Accepted: 07/07/2011] [Indexed: 12/31/2022] Open
Abstract
Background CGGBP1 is a CGG-triplet repeat binding protein, which affects transcription from CGG-triplet-rich promoters such as the FMR1 gene and the ribosomal RNA gene clusters. Earlier, we reported some previously unknown functions of CGGBP1 in gene expression during heat shock stress response. Recently we had found CGGBP1 to be a cell cycle regulatory midbody protein required for normal cytokinetic abscission in normal human fibroblasts, which have all the cell cycle regulatory mechanisms intact. Results In this study we explored the role of CGGBP1 in the cell cycle in various cancer cell lines. CGGBP1 depletion by RNA interference in tumor-derived cells caused an increase in the cell population at G0/G1 phase and reduced the number of cells in the S phase. CGGBP1 depletion also increased the expression of cell cycle regulatory genes CDKN1A and GAS1, associated with reductions in histone H3 lysine 9 trimethylation in their promoters. By combining RNA interference and genetic mutations, we found that the role of CGGBP1 in cell cycle involves multiple mechanisms, as single deficiencies of CDKN1A, GAS1 as well as TP53, INK4A or ARF failed to rescue the G0/G1 arrest caused by CGGBP1 depletion. Conclusions Our results show that CGGBP1 expression is important for cell cycle progression through multiple parallel mechanisms including the regulation of CDKN1A and GAS1 levels.
Collapse
Affiliation(s)
- Umashankar Singh
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, 751 85 Uppsala, Sweden.
| | | | | | | |
Collapse
|
39
|
Landreville S, Vigneault F, Bergeron MA, Leclerc S, Gaudreault M, Morcos M, Mouriaux F, Salesse C, Guérin SL. Suppression of α5 gene expression is closely related to the tumorigenic properties of uveal melanoma cell lines. Pigment Cell Melanoma Res 2011; 24:643-55. [PMID: 21592318 DOI: 10.1111/j.1755-148x.2011.00869.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Cancer aggressiveness is related to the ability of cancer cells to escape the anchorage dependency toward the extracellular matrix, a process regulated by the integrin α5β1 and its ligand fibronectin. Here, we characterized the expression of the α5 gene in human uveal melanoma cell lines with distinct tumorigenic properties and investigated some of the mechanisms underlying the variations of their malignancy. Strong and weak expression of α5 was observed in cells with no (T108/T115) and high (T97/T98) tumorigenic properties, respectively. Expression and DNA binding of the transcription factors Sp1, activator protein 1 (AP-1) (both acting as activators), and nuclear factor I (NFI) (a strong repressor) to the α5 promoter were demonstrated in all cell lines. A reduced expression of AP-1 combined with a dramatic increase in NFI correlated with the suppression of α5 expression in T97 and T98 cells. Restoring α5 expression in T97 cells entirely abolished their tumorigenicity in immunodeficient mice. These uveal melanoma cell lines might therefore prove particularly useful as cellular models to investigate α5β1 function in the pathogenesis of invasive uveal melanoma.
Collapse
Affiliation(s)
- Solange Landreville
- Department of Ophthalmology & Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Pjanic M, Pjanic P, Schmid C, Ambrosini G, Gaussin A, Plasari G, Mazza C, Bucher P, Mermod N. Nuclear factor I revealed as family of promoter binding transcription activators. BMC Genomics 2011; 12:181. [PMID: 21473784 PMCID: PMC3082249 DOI: 10.1186/1471-2164-12-181] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2009] [Accepted: 04/07/2011] [Indexed: 12/11/2022] Open
Abstract
Background Multiplex experimental assays coupled to computational predictions are being increasingly employed for the simultaneous analysis of many specimens at the genome scale, which quickly generates very large amounts of data. However, inferring valuable biological information from the comparisons of very large genomic datasets still represents an enormous challenge. Results As a study model, we chose the NFI/CTF family of mammalian transcription factors and we compared the results obtained from a genome-wide study of its binding sites with chromatin structure assays, gene expression microarray data, and in silico binding site predictions. We found that NFI/CTF family members preferentially bind their DNA target sites when they are located around transcription start sites when compared to control datasets generated from the random subsampling of the complete set of NFI binding sites. NFI proteins preferably associate with the upstream regions of genes that are highly expressed and that are enriched in active chromatin modifications such as H3K4me3 and H3K36me3. We postulate that this is a causal association and that NFI proteins mainly act as activators of transcription. This was documented for one member of the family (NFI-C), which revealed as a more potent gene activator than repressor in global gene expression analysis. Interestingly, we also discovered the association of NFI with the tri-methylation of lysine 9 of histone H3, a chromatin marker previously associated with the protection against silencing of telomeric genes by NFI. Conclusion Taken together, we illustrate approaches that can be taken to analyze large genomic data, and provide evidence that NFI family members may act in conjunction with specific chromatin modifications to activate gene expression.
Collapse
Affiliation(s)
- Milos Pjanic
- Institute of Biotechnology, University of Lausanne, Lausanne, Switzerland.
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Singh SK, Wilczynska KM, Grzybowski A, Yester J, Osrah B, Bryan L, Wright S, Griswold-Prenner I, Kordula T. The unique transcriptional activation domain of nuclear factor-I-X3 is critical to specifically induce marker gene expression in astrocytes. J Biol Chem 2010; 286:7315-26. [PMID: 21189253 DOI: 10.1074/jbc.m110.152421] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Transcription factors of the nuclear factor 1 (NFI) family regulate normal brain development in vertebrates. However, multiple splice variants of four NFI isoforms exist, and their biological functions have yet to be elucidated. Here, we cloned and analyzed human NFI-X3, a novel splice variant of the nfix gene, which contains a unique transcriptional activation (TA) domain completely conserved in primates. In contrast to previously cloned NFI-X1, overexpression of NFI-X3 potently activates NFI reporters, including glial fibrillary acidic protein (GFAP) reporter, in astrocytes and glioma cells. The GAL4 fusion protein containing the TA domain of NFI-X3 strongly activates the GAL4 reporter, whereas the TA domain of NFI-X1 is ineffective. The expression of NFI-X3 is dramatically up-regulated during the differentiation of neural progenitors to astrocytes and precedes the expression of astrocyte markers, such as GFAP and SPARCL1 (Secreted Protein, Acidic and Rich in Cysteines-like 1). Overexpression of NFI-X3 dramatically up-regulates GFAP and SPARCL1 expression in glioma cells, whereas the knockdown of NFI-X3 diminishes the expression of both GFAP and SPARCL1 in astrocytes. Although activation of astrocyte-specific genes involves DNA demethylation and subsequent increase of histone acetylation, NFI-X3 activates GFAP expression, in part, by inducing alterations in the nucleosome architecture that lead to the increased recruitment of RNA polymerase II.
Collapse
Affiliation(s)
- Sandeep K Singh
- Department of Biochemistry, Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia 23298, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Lichner Z, Páll E, Kerekes A, Pállinger E, Maraghechi P, Bosze Z, Gócza E. The miR-290-295 cluster promotes pluripotency maintenance by regulating cell cycle phase distribution in mouse embryonic stem cells. Differentiation 2010; 81:11-24. [PMID: 20864249 DOI: 10.1016/j.diff.2010.08.002] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2009] [Revised: 07/13/2010] [Accepted: 08/12/2010] [Indexed: 12/23/2022]
Abstract
The mmu-miR-290-295 cluster codes for a family of microRNAs (miRNAs) that are expressed de novo during early embryogenesis and are specific for mouse embryonic stem cells (ESC) and embryonic carcinoma cells (ECC). Detailed sequence analysis and alignment studies of miR-290-295 precursors demonstrated that the cluster has evolved by repeated duplication events of the ancient miR-290 precursor. We show that under serum starvation, overexpression of miR-290-295 miRNAs withhold ES cells from early differentiation, ensures their high proliferation rate and capacity for forming alkaline phosphate positive colonies. Transcriptome analysis revealed that differentiation related marker genes are underexpressed upon high miR-290-295 level. Importantly, miR-290-295 overexpression prevents ES cells from accumulation in G1 phase at low serum level, and seems to regulate cell cycle in different phases. Our data underline that miR-290-295 miRNAs contribute to the natural absence of G1 checkpoint in embryonic stem cells. We define the cell cycle regulators Wee1 and Fbxl5 as potential direct targets of miR-290-295 miRNAs in vitro. Our results suggest that miR-290-295 miRNAs exhibit their effect predominantly through the regulation of cell cycle phase distribution.
Collapse
Affiliation(s)
- Zsuzsanna Lichner
- Agricultural Biotechnology Center, H-2100, Szent-Györgyi A, Str. 4, Gödöllő, Hungary
| | | | | | | | | | | | | |
Collapse
|
43
|
Plasari G, Edelmann S, Högger F, Dusserre Y, Mermod N, Calabrese A. Nuclear factor I-C regulates TGF-{beta}-dependent hair follicle cycling. J Biol Chem 2010; 285:34115-25. [PMID: 20729551 DOI: 10.1074/jbc.m110.120659] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Skin appendages such as teeth and hair share several common signaling pathways. The nuclear factor I C (NFI-C) transcription factor has been implicated in tooth development, but a potential role in hair growth had not been assessed. In this study we found that NFI-C regulates the onset of the hair growth cycle. NFI-C(-/-) mice were delayed in the transition from the telogen to anagen phase of the hair follicle cycle after either experimental depilation or spontaneous hair loss. Lack of NFI-C resulted in delayed induction of the sonic hedgehog, Wnt5a, and Lef1 gene expression, which are key regulators of the hair follicle growth initiation. NFI-C(-/-) mice also showed elevated levels of transforming growth factor β1 (TGF-β1), an inhibitor of keratinocyte proliferation, and of the cell cycle inhibitor p21 at telogen. Reduced expression of Ki67, a marker of cell proliferation, was noted at the onset of anagen, indicating impaired activation of the hair progenitor cells. These findings implicate NFI-C in the repression of TGF-β1 signaling during telogen stage, resulting in the delay of progenitor cell proliferation and hair follicle regeneration in NFI-C-deficient mice. Taken together with prior observations, these findings also designate NFI-C as a regulator of adult progenitor cell proliferation and of postnatal tissue growth or regeneration.
Collapse
Affiliation(s)
- Genta Plasari
- Institute of Biotechnology, University of Lausanne, 1015 Lausanne, Switzerland
| | | | | | | | | | | |
Collapse
|
44
|
Nassiri M, Liu J, Kulak S, Uwiera RRE, Aird WC, Ballermann BJ, Jahroudi N. Repressors NFI and NFY participate in organ-specific regulation of von Willebrand factor promoter activity in transgenic mice. Arterioscler Thromb Vasc Biol 2010; 30:1423-9. [PMID: 20431063 DOI: 10.1161/atvbaha.110.206680] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE To determine the role of repressors in cell type and organ-specific activation of von Willebrand factor (VWF) promoter sequences -487 to 247 in vivo. METHODS AND RESULTS Activation patterns of wild-type and mutant VWF promoters (sequences -487 to 247) containing mutations in repressors nuclear factor-I (NFI)- and nuclear factor Y (NFY)-binding sites were analyzed in transgenic mice. Mutation of the NFI-binding site activated the promoter in heart and lung endothelial cells, whereas mutation of the NFY-binding site activated the promoter in kidney vasculature. Immunofluorescence analyses showed that NFIB was predominant in heart and lung endothelial cells, whereas NFIX was predominantly detected in kidney endothelial cell nuclei. By using chromatin immunoprecipitation, we demonstrated that the distal lung-specific enhancer (containing a YY1 site) of the VWF gene is brought in proximity to the NFI binding site. CONCLUSIONS The NFI and NFY repressors contribute differentially to organ-specific regulation of the VWF promoter, and the organ-specific action of NFI may reflect its organ-specific isoform distribution. In addition, the lung-specific enhancer region of the endogenous VWF gene may inhibit NFI repressor function through chromatin looping, which can approximate the 2 regions.
Collapse
Affiliation(s)
- Marjan Nassiri
- Department of Medicine, University of Alberta, Edmonton, AL, Canada
| | | | | | | | | | | | | |
Collapse
|
45
|
Robitaille H, Simard-Bisson C, Larouche D, Tanguay RM, Blouin R, Germain L. The Small Heat-Shock Protein Hsp27 Undergoes ERK-Dependent Phosphorylation and Redistribution to the Cytoskeleton in Response to Dual Leucine Zipper-Bearing Kinase Expression. J Invest Dermatol 2010; 130:74-85. [DOI: 10.1038/jid.2009.185] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
46
|
Millau JF, Bastien N, Bouchard EF, Drouin R. p53 Pre- and post-binding event theories revisited: stresses reveal specific and dynamic p53-binding patterns on the p21 gene promoter. Cancer Res 2009; 69:8463-71. [PMID: 19843844 DOI: 10.1158/0008-5472.can-09-2036] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
p53 is a master transcription factor that prevents neoplasia and genomic instability. It is an important target for anticancer drug design. Understanding the molecular mechanisms behind its transcriptional activities in normal cells is a prerequisite to further understand the deregulation effected by mutant p53 in cancerous cells. Currently, how p53 coordinates transcription programs in response to stress remains unclear. One theory proposes that stresses induce pre-binding events that direct p53 to bind to specific response elements, whereas a second posits that, in response to stress, p53 binds most response elements and post-binding events then regulate transcription initiation. It is critical to establish the relevance of both theories and investigate whether stresses induce specific p53-binding patterns correlated with effector gene induction. Using unique in cellulo genomic footprinting experiments, we studied p53 binding to the five response elements of p21 in response to stresses and monitored p21 mRNA variant transcription. We show clear footprints of p53 bound to response elements in living cells and reveal that the binding of p53 to response elements is transient, subject to dynamic changes during stress responses, and influenced by response element pentamer orientations. We show further that stresses lead to specific p53-binding patterns correlated with particular p21 mRNA variant transcription profiles and that p53 binding is necessary but not sufficient to induce p21 transcription. Our results indicate that pre- and post-binding events act together to regulate adapted stress responses; this paves the way to the unification of pre- and post-binding event theories.
Collapse
Affiliation(s)
- Jean-François Millau
- Division of Genetics, Department of Pediatrics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | | | | | | |
Collapse
|
47
|
Lee DS, Park JT, Kim HM, Ko JS, Son HH, Gronostajski RM, Cho MI, Choung PH, Park JC. Nuclear factor I-C is essential for odontogenic cell proliferation and odontoblast differentiation during tooth root development. J Biol Chem 2009; 284:17293-17303. [PMID: 19386589 DOI: 10.1074/jbc.m109.009084] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Our previous studies have demonstrated that nuclear factor I-C (NFI-C) null mice developed short molar roots that contain aberrant odontoblasts and abnormal dentin formation. Based on these findings, we performed studies to elucidate the function of NFI-C in odontoblasts. Initial studies demonstrated that aberrant odontoblasts become dissociated and trapped in an osteodentin-like mineralized tissue. Abnormal odontoblasts exhibit strong bone sialoprotein expression but a decreased level of dentin sialophosphoprotein expression when compared with wild type odontoblasts. Loss of Nfic results in an increase in p-Smad2/3 expression in aberrant odontoblasts and pulp cells in the subodontoblastic layer in vivo and primary pulp cells from Nfic-deficient mice in vitro. Cell proliferation analysis of both cervical loop and ectomesenchymal cells of the Nfic-deficient mice revealed significantly decreased proliferative activity compared with wild type mice. In addition, Nfic-deficient primary pulp cells showed increased expression of p21 and p16 but decreased expression of cyclin D1 and cyclin B1, strongly suggesting cell growth arrest caused by a lack of Nfic activity. Analysis of the pulp and abnormal dentin in Nfic-deficient mice revealed an increase in apoptotic activity. Further, Nfic-deficient primary pulp cells exhibited an increase in caspase-8 and -3 activation, whereas the cleaved form of Bid was hardly detected. These results indicate that the loss of Nfic leads to the suppression of odontogenic cell proliferation and differentiation and induces apoptosis of aberrant odontoblasts during root formation, thereby contributing to the formation of short roots.
Collapse
Affiliation(s)
- Dong-Seol Lee
- From the Department of Oral Histology-Developmental Biology, Dental Research Institute, Seoul 110-749, Korea
| | - Jong-Tae Park
- From the Department of Oral Histology-Developmental Biology, Dental Research Institute, Seoul 110-749, Korea
| | - Hyun-Man Kim
- From the Department of Oral Histology-Developmental Biology, Dental Research Institute, Seoul 110-749, Korea
| | - Jea Seung Ko
- From the Department of Oral Histology-Developmental Biology, Dental Research Institute, Seoul 110-749, Korea
| | - Ho-Hyun Son
- Conservative Dentistry, Seoul 110-749, Korea
| | - Richard M Gronostajski
- Department of Biochemistry and the Program in Neuroscience, School of Medicine and Biomedical Science, Buffalo, New York 14214-3092
| | - Moon-Il Cho
- Department of Oral Biology, School of Dental Medicine, State University of New York at Buffalo, Buffalo, New York 14214-3092
| | - Pill-Hoon Choung
- Oral & Maxillofacial Surgery, Seoul 110-749, Korea; Tooth Bioengineering National Research Lab, School of Dentistry, Seoul National University, Seoul 110-749, Korea
| | - Joo-Cheol Park
- From the Department of Oral Histology-Developmental Biology, Dental Research Institute, Seoul 110-749, Korea.
| |
Collapse
|
48
|
Singh U, Bongcam-Rudloff E, Westermark B. A DNA sequence directed mutual transcription regulation of HSF1 and NFIX involves novel heat sensitive protein interactions. PLoS One 2009; 4:e5050. [PMID: 19337383 PMCID: PMC2660424 DOI: 10.1371/journal.pone.0005050] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2009] [Accepted: 03/05/2009] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Though the Nuclear factor 1 family member NFIX has been strongly implicated in PDGFB-induced glioblastoma, its molecular mechanisms of action remain unknown. HSF1, a heat shock-related transcription factor is also a powerful modifier of carcinogenesis by several factors, including PDGFB. How HSF1 transcription is controlled has remained largely elusive. METHODOLOGY/PRINCIPAL FINDINGS By combining microarray expression profiling and a yeast-two-hybrid screen, we identified that NFIX and its interactions with CGGBP1 and HMGN1 regulate expression of HSF1. We found that CGGBP1 organizes a bifunctional transcriptional complex at small CGG repeats in the HSF1 promoter. Under chronic heat shock, NFIX uses CGGBP1 and HMGN1 to get recruited to this promoter and in turn affects their binding to DNA. Results show that the interactions of NFIX with CGGBP1 and HMGN1 in the soluble fraction are heat shock sensitive due to preferential localization of CGGBP1 to heterochromatin after heat shock. HSF1 in turn was found to bind to the NFIX promoter and repress its expression in a heat shock sensitive manner. CONCLUSIONS/SIGNIFICANCE NFIX and HSF1 exert a mutual transcriptional repressive effect on each other which requires CGG repeat in HSF1 promoter and HSF1 binding site in NFIX promoter. We unravel a unique mechanism of heat shock sensitive DNA sequence-directed reciprocal transcriptional regulation between NFIX and HSF1. Our findings provide new insights into mechanisms of transcription regulation under stress.
Collapse
Affiliation(s)
- Umashankar Singh
- Department of Genetics and Pathology, Uppsala University, Uppsala, Sweden
- * E-mail: (US); (BW)
| | | | - Bengt Westermark
- Department of Genetics and Pathology, Uppsala University, Uppsala, Sweden
- * E-mail: (US); (BW)
| |
Collapse
|
49
|
Pérez-Casellas LA, Wang X, Howard KD, Rehage MW, Strong DD, Linkhart TA. Nuclear factor I transcription factors regulate IGF binding protein 5 gene transcription in human osteoblasts. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2008; 1789:78-87. [PMID: 18809517 DOI: 10.1016/j.bbagrm.2008.08.013] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2008] [Revised: 08/11/2008] [Accepted: 08/26/2008] [Indexed: 11/17/2022]
Abstract
Insulin-like growth factor binding protein 5 (IGFBP5) is expressed in many cell types including osteoblasts and modulates IGF activities. IGFBP5 may affect osteoblasts and bone formation, in part by mechanisms independent of binding IGFs. The highly conserved IGFBP5 proximal promoter within 100 nucleotides of the start of transcription contains functional cis regulatory elements for C/EBP, Myb and AP-2. We report evidence for a functional Nuclear Factor I (NFI) cis element that mediates activation or repression of IGFBP5 transcription by the NFI gene family. All four NFI genes were expressed in human osteoblast cultures and osteosarcoma cell lines. Co-transfection with human IGFBP5 promoter luciferase reporter and murine Nfi expression vectors showed that Nfib was the most active in stimulating transcription. Nfix was less active and Nfia and Nfic were inhibitory. Knockdown of NFIB and NFIC expression using siRNA decreased and increased IGFBP5 expression, respectively. Analysis of IGFBP5 promoter deletion and mutation reporter constructs identified a functional NFI cis element. All four NFI proteins bound the NFI site in electrophoretic mobility shift experiments and NFIB bound in chromatin immunoprecipitation assays. Results suggest that NFI proteins are important regulators of IGFBP5 expression in human osteoblasts and thus in modulating IGFBP5 functions in bone.
Collapse
|
50
|
Nasri S, More H, Graziano F, Ruzzo A, Wilson E, Dunbier A, McKinney C, Merriman T, Guilford P, Magnani M, Humar B. A novel diffuse gastric cancer susceptibility variant in E-cadherin (CDH1) intron 2: a case control study in an Italian population. BMC Cancer 2008; 8:138. [PMID: 18482459 PMCID: PMC2412889 DOI: 10.1186/1471-2407-8-138] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2007] [Accepted: 05/15/2008] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Inherited genetic factors such as E-cadherin (CDH1) promoter variants are believed to influence the risk towards sporadic diffuse gastric cancer (DGC). Recently, a new regulatory region essential for CDH1 transcription has been identified in CDH1 intron 2. METHODS We genotyped all known polymorphisms located within conserved sequences of CDH1 intron 2 (rs10673765, rs9932686, rs1125557, rs9282650, rs9931853) in an Italian population consisting of 134 DGC cases and 100 healthy controls (55 patient relatives and 45 unrelated, matched individuals). The influence of individual variants on DGC risk was assessed using chi2-tests and logistic regression. The relative contribution of alleles was estimated by haplotype analysis. RESULTS We observed a significant (p < 0.0004) association of the CDH1 163+37235G>A variant (rs1125557) with DGC risk. Odds ratios were 4.55 (95%CI = 2.09-9.93) and 1.38 (95%CI = 0.75-2.55) for AA and GA carriers, respectively. When adjusted for age, sex, smoking status, alcohol intake and H. pylori infection, the risk estimates remained largely significant for AA carriers. Haplotype analysis suggested the 163+37235A-allele contributes to disease risk independently of the other variants studied. CONCLUSION The CDH1 163+37235G>A polymorphism may represent a novel susceptibility variant for sporadic DGC if confirmed in other populations. Considering the broad expression of E-cadherin in epithelia, this exploratory study encourages further evaluation of the 163+37235A-allele as a susceptibility variant in other carcinomas.
Collapse
Affiliation(s)
- Soroush Nasri
- Cancer Genetics Laboratory, Department of Biochemistry, University of Otago, Dunedin 9054, Aotearoa New Zealand.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|