1
|
Qi M, Sun LA, Zheng LR, Zhang J, Han YL, Wu F, Zhao J, Niu WH, Fei MX, Jiang XC, Zhou ML. Expression and potential role of FOSB in glioma. Front Mol Neurosci 2022; 15:972615. [PMID: 36311014 PMCID: PMC9597691 DOI: 10.3389/fnmol.2022.972615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Accepted: 09/08/2022] [Indexed: 11/17/2022] Open
Abstract
Background FOSB is reported to be an oncogene in a variety of tumors. However, the expression and role of FOSB in glioma remain obscure. In this study, we aimed to explore the expression of FOSB in glioma and its biological role in glioblastoma multiforme (GBM). Methods Western blot, immunohistochemical staining, and quantitative real-time polymerase chain reaction (RT-qPCR) were used to detect the expression of FOSB in clinical samples. FOSB was knocked down in cells to determine the effects of FOSB on the phenotypic changes of tumors by plate cloning, CCK-8 assay, and Transwell assay. Finally, subcutaneous tumorigenesis in nude mice was used to observe the tumorigenesis of glioma cell lines after the knockdown of the FOSB gene. Results FOSB expression was higher in glioma compared with normal brain tissue. After the downregulation of FOSB, the expression of cleaved caspase-3 increased. Plate cloning and CCK-8 experiments showed that the proliferation of glioma cell lines decreased. The Transwell assay demonstrated that the glioblastoma cell lines had lower migration ability after the knockdown of FOSB. Finally, the tumor volume of U87 glioma cells in group sh-FOSB was smaller than that in the control group. The TUNEL staining in vitro showed that the apoptosis of sh-FOSB glioma cells increased. Conclusion FOSB was highly expressed in glioma tissues. The viability of glioma cells decreased, and the ability of glioma cells to proliferate and migrate was reduced when FOSB was downregulated. Hence, FOSB may promote the development and migration of gliomas.
Collapse
Affiliation(s)
- Min Qi
- The Translational Research Institute for Neurological Disorders of Wannan Medical College, Department of Neurosurgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, China
| | - Le-an Sun
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Lan-rong Zheng
- Department of Pathology, Wannan Medical College, Wuhu, China
| | - Jia Zhang
- The Translational Research Institute for Neurological Disorders of Wannan Medical College, Department of Neurosurgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, China
| | - Yan-ling Han
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Feng Wu
- Department of Anatomy, Wannan Medical College, Wuhu, China
| | - Jian Zhao
- Department of Anatomy, Wannan Medical College, Wuhu, China
| | - Wen-hao Niu
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Mao-xing Fei
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Xiao-chun Jiang
- The Translational Research Institute for Neurological Disorders of Wannan Medical College, Department of Neurosurgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, China
- Xiao-chun Jiang
| | - Meng-liang Zhou
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
- *Correspondence: Meng-liang Zhou
| |
Collapse
|
2
|
Aloizou AM, Pateraki G, Siokas V, Mentis AFA, Liampas I, Lazopoulos G, Kovatsi L, Mitsias PD, Bogdanos DP, Paterakis K, Dardiotis E. The role of MiRNA-21 in gliomas: Hope for a novel therapeutic intervention? Toxicol Rep 2020; 7:1514-1530. [PMID: 33251119 PMCID: PMC7677650 DOI: 10.1016/j.toxrep.2020.11.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 10/30/2020] [Accepted: 11/02/2020] [Indexed: 12/20/2022] Open
Abstract
Gliomas are the most common primary brain tumors in adults. They are generally very resistant to treatment and are therefore associated with negative outcomes. MicroRNAs (miRNAs) are small, non-coding RNA molecules that affect many cellular processes by regulating gene expression and, post-transcriptionally, the translation of mRNAs. MiRNA-21 has been consistently shown to be upregulated in glioma and research has shown that it is involved in a wide variety of biological pathways, promoting tumor cell survival and invasiveness. Furthermore, it has been implicated in resistance to treatment, both against chemotherapy and radiotherapy. In this review, we gathered the existent data on miRNA-21 and gliomas, in terms of its expression levels, association with grade and prognosis, the pathways it involves and its targets in glioma, and finally how it leads to treatment resistance. Furthermore, we discuss how this knowledge could be applied in clinical practice in the years to come. To our knowledge, this is the first review to assess in extent and depth the role of miRNA-21 in gliomas.
Collapse
Affiliation(s)
- Athina-Maria Aloizou
- Department of Neurology, Laboratory of Neurogenetics, University of Thessaly, University Hospital of Larissa, Larissa, Greece
| | - Georgia Pateraki
- Department of Neurology, Laboratory of Neurogenetics, University of Thessaly, University Hospital of Larissa, Larissa, Greece
| | - Vasileios Siokas
- Department of Neurology, Laboratory of Neurogenetics, University of Thessaly, University Hospital of Larissa, Larissa, Greece
| | - Alexios-Fotios A Mentis
- Department of Neurology, Laboratory of Neurogenetics, University of Thessaly, University Hospital of Larissa, Larissa, Greece.,Public Health Laboratories, Hellenic Pasteur Institute, Athens, Greece
| | - Ioannis Liampas
- Department of Neurology, Laboratory of Neurogenetics, University of Thessaly, University Hospital of Larissa, Larissa, Greece
| | - George Lazopoulos
- Department of Cardiothoracic Surgery, University General Hospital of Heraklion, Medical School, University of Crete, 71003 Heraklion, Greece
| | - Leda Kovatsi
- Laboratory of Forensic Medicine and Toxicology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki 54124, Greece
| | - Panayiotis D Mitsias
- Department of Neurology, School of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Dimitrios P Bogdanos
- Department of Rheumatology and Clinical Immunology, University General Hospital of Larissa, Faculty of Medicine, School of Health Sciences, University of Thessaly, Viopolis 40500, Larissa, Greece
| | - Konstantinos Paterakis
- Department of Neurosurgery, University of Thessaly, University Hospital of Larissa, Larissa, Greece
| | - Efthimios Dardiotis
- Department of Neurology, Laboratory of Neurogenetics, University of Thessaly, University Hospital of Larissa, Larissa, Greece
| |
Collapse
|
3
|
Ayanlaja AA, Zhang B, Ji G, Gao Y, Wang J, Kanwore K, Gao D. The reversible effects of glial cell line-derived neurotrophic factor (GDNF) in the human brain. Semin Cancer Biol 2018; 53:212-222. [PMID: 30059726 DOI: 10.1016/j.semcancer.2018.07.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Revised: 07/10/2018] [Accepted: 07/18/2018] [Indexed: 12/20/2022]
Abstract
Glial cell line-derived neurotrophic factor (GDNF) is a potent survival factor, and a member of the transforming growth factor β (TGF-β) superfamily acting on different neuronal activities. GDNF was originally identified as a neurotrophic factor crucially involved in the survival of dopaminergic neurons of the nigrostriatal pathway and is currently an established therapeutic target in Parkinson's disease. However, GDNF was later reported to be highly expressed in gliomas, especially in glioblastomas, and was demonstrated as a potent proliferation factor involved in the development and migration of gliomas. Here, we review our current understanding and progress made so far by researchers in our laboratories with references to relevant articles to support our discoveries. We present past and recent discoveries on the mechanisms involved in the protection of neurons by GDNF and examine its emerging roles in gliomas, as well as reasons for the abnormal expression in Glioblastoma Multiforme (GBM). Collectively, our work establishes a paradigm by which the ability of GDNF to protect dopaminergic neurons from degradation and its corresponding effects on glioma cells points to an underlying biological vulnerability in the effects of GDNF in the normal brain which can be subverted for use by cancer cells. Hence, presenting novel opportunities for intervention in glioma therapies.
Collapse
Affiliation(s)
- Abiola Abdulrahman Ayanlaja
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Baole Zhang
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - GuangQuan Ji
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Yue Gao
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Jie Wang
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Kouminin Kanwore
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - DianShuai Gao
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China.
| |
Collapse
|
4
|
Sideek MA, Teia A, Kopecki Z, Cowin AJ, Gibson MA. Co-localization of LTBP-2 with FGF-2 in fibrotic human keloid and hypertrophic scar. J Mol Histol 2015; 47:35-45. [PMID: 26644005 DOI: 10.1007/s10735-015-9645-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 11/25/2015] [Indexed: 02/06/2023]
Abstract
We have recently shown that Latent transforming growth factor-beta-1 binding protein-2 (LTBP-2) has a single high-affinity binding site for fibroblast growth factor-2 (FGF-2) and that LTBP-2 blocks FGF-2 induced cell proliferation. Both proteins showed strong co-localisation within keloid skin from a single patient. In the current study, using confocal microscopy, we have investigated the distribution of the two proteins in normal and fibrotic skin samples including normal scar tissue, hypertrophic scars and keloids from multiple patients. Consistently, little staining for either protein was detected in normal adult skin and normal scar samples but extensive co-localisation of the two proteins was observed in multiple examples of hypertrophic scars and keloids. LTBP-2 and FGF-2 were co-localised to fine fibrous elements within the extracellular matrix identified as elastic fibres by immunostaining with anti-fibrillin-1 and anti-elastin antibodies. Furthermore, qPCR analysis of RNA samples from multiple patients confirmed dramatically increased expression of LTBP-2 and FGF-2, similar TGF-beta 1, in hypertrophic scar compared to normal skin and scar tissue. Overall the results suggest that elevated LTBP-2 may bind and sequester FGF-2 on elastic fibres in fibrotic tissues and modulate FGF-2's influence on the repair and healing processes.
Collapse
Affiliation(s)
- Mohamed A Sideek
- Discipline of Anatomy and Pathology, School of Medicine, University of Adelaide, Adelaide, SA, 5005, Australia.,Department of Biomedical Sciences, Kulliyyah of Allied Health Sciences, International Islamic University Malaysia, 25200, Kuantan, Pahang, Malaysia
| | - Abdulrahman Teia
- Discipline of Anatomy and Pathology, School of Medicine, University of Adelaide, Adelaide, SA, 5005, Australia
| | - Zlatko Kopecki
- Regenerative Medicine, Mawson Institute, University of South Australia, Adelaide, SA, 5095, Australia
| | - Allison J Cowin
- Regenerative Medicine, Mawson Institute, University of South Australia, Adelaide, SA, 5095, Australia
| | - Mark A Gibson
- Discipline of Anatomy and Pathology, School of Medicine, University of Adelaide, Adelaide, SA, 5005, Australia.
| |
Collapse
|
5
|
LTBP-2 Has a Single High-Affinity Binding Site for FGF-2 and Blocks FGF-2-Induced Cell Proliferation. PLoS One 2015; 10:e0135577. [PMID: 26263555 PMCID: PMC4532469 DOI: 10.1371/journal.pone.0135577] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2014] [Accepted: 07/24/2015] [Indexed: 12/18/2022] Open
Abstract
Latent transforming growth factor-beta-1 binding protein-2 (LTBP-2) belongs to the fibrillin-LTBP superfamily of extracellular matrix proteins. LTBPs and fibrillins are involved in the sequestration and storage of latent growth factors, particularly transforming growth factor β (TGF-β), in tissues. Unlike other LTBPs, LTBP-2 does not covalently bind TGF-β, and its molecular functions remain unclear. We are screening LTBP-2 for binding to other growth factors and have found very strong saturable binding to fibroblast growth factor-2 (FGF-2) (Kd = 1.1 nM). Using a series of recombinant LTBP-2 fragments a single binding site for FGF-2 was identified in a central region of LTBP-2 consisting of six tandem epidermal growth factor-like (EGF-like) motifs (EGFs 9–14). This region was also shown to contain a heparin/heparan sulphate-binding site. FGF-2 stimulation of fibroblast proliferation was completely negated by the addition of 5-fold molar excess of LTBP-2 to the assay. Confocal microscopy showed strong co-localisation of LTBP-2 and FGF-2 in fibrotic keloid tissue suggesting that the two proteins may interact in vivo. Overall the study indicates that LTBP-2 is a potent inhibitor of FGF-2 that may influence FGF-2 bioactivity during wound repair particularly in fibrotic tissues.
Collapse
|
6
|
Kajitani N, Hisaoka-Nakashima K, Okada-Tsuchioka M, Hosoi M, Yokoe T, Morioka N, Nakata Y, Takebayashi M. Fibroblast growth factor 2 mRNA expression evoked by amitriptyline involves extracellular signal-regulated kinase-dependent early growth response 1 production in rat primary cultured astrocytes. J Neurochem 2015; 135:27-37. [DOI: 10.1111/jnc.13247] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Revised: 07/08/2015] [Accepted: 07/09/2015] [Indexed: 01/17/2023]
Affiliation(s)
- Naoto Kajitani
- Department of Pharmacology, Graduate School of Biomedical & Health Sciences; Hiroshima University; Minami-ku Hiroshima Japan
- Division of Psychiatry and Neuroscience; Institute for Clinical Research; National Hospital Organization (NHO) Kure Medical Center and Chugoku Cancer Center; Kure Japan
| | - Kazue Hisaoka-Nakashima
- Department of Pharmacology, Graduate School of Biomedical & Health Sciences; Hiroshima University; Minami-ku Hiroshima Japan
| | - Mami Okada-Tsuchioka
- Division of Psychiatry and Neuroscience; Institute for Clinical Research; National Hospital Organization (NHO) Kure Medical Center and Chugoku Cancer Center; Kure Japan
| | - Mayu Hosoi
- Division of Psychiatry and Neuroscience; Institute for Clinical Research; National Hospital Organization (NHO) Kure Medical Center and Chugoku Cancer Center; Kure Japan
| | - Toshiki Yokoe
- Department of Pharmacology, Graduate School of Biomedical & Health Sciences; Hiroshima University; Minami-ku Hiroshima Japan
| | - Norimitsu Morioka
- Department of Pharmacology, Graduate School of Biomedical & Health Sciences; Hiroshima University; Minami-ku Hiroshima Japan
| | - Yoshihiro Nakata
- Department of Pharmacology, Graduate School of Biomedical & Health Sciences; Hiroshima University; Minami-ku Hiroshima Japan
| | - Minoru Takebayashi
- Division of Psychiatry and Neuroscience; Institute for Clinical Research; National Hospital Organization (NHO) Kure Medical Center and Chugoku Cancer Center; Kure Japan
- Department of Psychiatry; National Hospital Organization (NHO) Kure Medical Center and Chugoku Cancer Center; Kure Japan
| |
Collapse
|
7
|
Christofides A, Kosmopoulos M, Piperi C. Pathophysiological mechanisms regulated by cytokines in gliomas. Cytokine 2014; 71:377-84. [PMID: 25458967 DOI: 10.1016/j.cyto.2014.09.008] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Revised: 09/17/2014] [Accepted: 09/25/2014] [Indexed: 12/16/2022]
Abstract
Glioma, a neuroglia originated malignancy, consists of one of the most aggressive primary tumors of the central nervous system with poor prognosis and lack of efficient treatment strategy. Cytokines have been implicated in several stages of glioma progression, participating in tumor onset, growth enhancement, angiogenesis and aggressiveness. Interestingly, cytokines have also the ability to inhibit glioma growth upon specific regulation or interplay with other molecules. This review addresses the dual role of major cytokines implicated in glioma pathology, pointing toward promising therapeutic approaches.
Collapse
Affiliation(s)
- Anthos Christofides
- Department of Biological Chemistry, University of Athens, Medical School, 11527 Athens, Greece
| | - Marinos Kosmopoulos
- Department of Biological Chemistry, University of Athens, Medical School, 11527 Athens, Greece
| | - Christina Piperi
- Department of Biological Chemistry, University of Athens, Medical School, 11527 Athens, Greece.
| |
Collapse
|
8
|
Ku MC, Wolf SA, Respondek D, Matyash V, Pohlmann A, Waiczies S, Waiczies H, Niendorf T, Synowitz M, Glass R, Kettenmann H. GDNF mediates glioblastoma-induced microglia attraction but not astrogliosis. Acta Neuropathol 2013; 125:609-20. [PMID: 23344256 DOI: 10.1007/s00401-013-1079-8] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Accepted: 01/09/2013] [Indexed: 12/14/2022]
Abstract
High-grade gliomas are the most common primary brain tumors. Their malignancy is promoted by the complex crosstalk between different cell types in the central nervous system. Microglia/brain macrophages infiltrate high-grade gliomas and contribute to their progression. To identify factors that mediate the attraction of microglia/macrophages to malignant brain tumors, we established a glioma cell encapsulation model that was applied in vivo. Mouse GL261 glioma cell line and human high-grade glioma cells were seeded into hollow fibers (HF) that allow the passage of soluble molecules but not cells. The glioma cell containing HF were implanted into one brain hemisphere and simultaneously HF with non-transformed fibroblasts (controls) were introduced into the contralateral hemisphere. Implanted mouse and human glioma- but not fibroblast-containing HF attracted microglia and up-regulated immunoreactivity for GFAP, which is a marker of astrogliosis. In this study, we identified GDNF as an important factor for microglial attraction: (1) GL261 and human glioma cells secret GDNF, (2) reduced GDNF production by siRNA in GL261 in mouse glioma cells diminished attraction of microglia, (3) over-expression of GDNF in fibroblasts promoted microglia attraction in our HF assay. In vitro migration assays also showed that GDNF is a strong chemoattractant for microglia. While GDNF release from human or mouse glioma had a profound effect on microglial attraction, the glioma-induced astrogliosis was not affected. Finally, we could show that injection of GL261 mouse glioma cells with GDNF knockdown by shRNA into mouse brains resulted in reduced tumor expansion and improved survival as compared to injection of control cells.
Collapse
Affiliation(s)
- Min-Chi Ku
- Department of Cellular Neuroscience, Max Delbrück Center for Molecular Medicine (MDC), Robert Rössle Str. 10, 13125 Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Gerhauser I, Hansmann F, Puff C, Kumnok J, Schaudien D, Wewetzer K, Baumgärtner W. Theiler's murine encephalomyelitis virus induced phenotype switch of microglia in vitro. J Neuroimmunol 2012; 252:49-55. [PMID: 22967352 DOI: 10.1016/j.jneuroim.2012.07.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2012] [Revised: 07/21/2012] [Accepted: 07/31/2012] [Indexed: 10/27/2022]
Abstract
The present in vitro study aimed to define the involvement of astrocytes and microglia in the initial inflammatory response of Theiler's murine encephalomyelitis (TME), a virus-induced mouse model of multiple sclerosis, and whether intralesional microglia exert pro- (M1) or anti-inflammatory (M2) effects following TME virus (TMEV) infection. Therefore astrocytes and microglia were purified from neonatal murine brains and inoculated either with TMEV or mock-solution. Gene expression of IL-1, IL-2, IL-10, IL-12, TNF, TNF receptors (TNFR1, TNFR2), TGFβ1, IFNγ and transcription factors NF-κB (p50, p65) and AP-1 (c-jun, c-fos) were quantified using RT-qPCR at 6, 48, and 240h post infection (hpi). In addition, IL-1, IL-10, IL-12, TNF and TGFβ1 mRNA transcripts were investigated at 168 hpi in TMEV- and mock-infected SJL/J mice. Overall in vitro astrocytes showed a significant higher amount of viral RNA compared to microglia. In addition, TMEV-infected astrocytes showed higher numbers of IL-1, IL-12 and TNF transcripts at 48 hpi. In microglia high IL-10 and low IL-12 mRNA levels were detected at 48 hpi, while the opposite was the case at 240 hpi. In addition, TNF mRNA was increased in microglia at 240 hpi. In addition, the observed up-regulation of IL-1, IL-12 and IL-10 in the early phase of TME in vivo substantiates the relevance of these cytokines during the disease induction. Summarized data indicate that TMEV infection of microglia induces a switch from the anti-inflammatory (M2) during the early phase to the pro-inflammatory (M1) phenotype in the later phase of the infection. The simultaneous expression of TNF and its receptors by both cell types might generate autocrine feedback loops possibly associated with pro-inflammatory actions of astrocytes via TNFR1.
Collapse
Affiliation(s)
- I Gerhauser
- Department of Pathology, University of Veterinary Medicine Hannover, Bünteweg 17, D-30559 Hannover, Germany.
| | | | | | | | | | | | | |
Collapse
|
10
|
Luo M, Liu Z, Chen G, Hao H, Lu T, Cui Y, Lei M, Verfaillie CM, Liu Z. High glucose enhances TGF-β1 expression in rat bone marrow stem cells via ERK1/2-mediated inhibition of STAT3 signaling. Life Sci 2012; 90:509-18. [DOI: 10.1016/j.lfs.2012.01.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2011] [Revised: 11/23/2011] [Accepted: 01/09/2012] [Indexed: 01/01/2023]
|
11
|
King MD, McCracken DJ, Wade FM, Meiler SE, Alleyne CH, Dhandapani KM. Attenuation of hematoma size and neurological injury with curcumin following intracerebral hemorrhage in mice. J Neurosurg 2011; 115:116-23. [PMID: 21417704 DOI: 10.3171/2011.2.jns10784] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
OBJECT Intracerebral hemorrhage (ICH) is associated with significant morbidity and mortality. Acute hematoma enlargement is an important predictor of neurological injury and poor clinical prognosis; but neurosurgical clot evacuation may not be feasible in all patients and treatment options remain largely supportive. Thus, novel therapeutic approaches to promote hematoma resolution are needed. In the present study, the authors investigated whether the curry spice curcumin limited neurovascular injury following ICH in mice. METHODS Intracerebral hemorrhage was induced in adult male CD-1 mice by intracerebral administration of collagenase or autologous blood. Clinically relevant doses of curcumin (75-300 mg/kg) were administered up to 6 hours after ICH, and hematoma volume, inflammatory gene expression, blood-brain barrier permeability, and brain edema were assessed over the first 72 hours. Neurological assessments were performed to correlate neurovascular protection with functional outcomes. RESULTS Curcumin increased hematoma resolution at 72 hours post-ICH. This effect was associated with a significant reduction in the expression of the proinflammatory mediators, tumor necrosis factor-α, interleukin-6, and interleukin-1β. Curcumin also reduced disruption of the blood-brain barrier and attenuated the formation of vasogenic edema following ICH. Consistent with the reduction in neuroinflammation and neurovascular injury, curcumin significantly improved neurological outcome scores after ICH. CONCLUSIONS Curcumin promoted hematoma resolution and limited neurological injury following ICH. These data may indicate clinical utility for curcumin as an adjunct therapy to reduce brain injury and improve patient outcome.
Collapse
Affiliation(s)
- Melanie D King
- Department of Neurosurgery, Georgia Health Sciences University, 1120 15th Street, Augusta, Georgia 30809, USA
| | | | | | | | | | | |
Collapse
|
12
|
Yum HY, Cho JY, Miller M, Broide DH. Allergen-induced coexpression of bFGF and TGF-β1 by macrophages in a mouse model of airway remodeling: bFGF induces macrophage TGF-β1 expression in vitro. Int Arch Allergy Immunol 2010; 155:12-22. [PMID: 21109744 DOI: 10.1159/000317213] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2009] [Accepted: 06/16/2010] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Basic fibroblast growth factor (bFGF) is a cytokine that is mitogenic for fibroblasts and smooth muscle and may play a role in airway remodeling in asthma. We have used a mouse model of chronic ovalbumin (OVA) allergen-induced airway remodeling to determine whether bFGF and fibroblast growth factor receptor-1 are expressed and regulated by corticosteroids in the airway, as well as to determine whether bFGF mediates expression of another proremodeling cytokine, transforming growth factor (TGF)-β1. METHODS The airway levels and localization of bFGF, FGF receptor-1 and TGF-β1 were determined by ELISA, immunohistology and image analysis in the remodeled airways of chronic OVA-challenged mice treated with either corticosteroids or diluent. In vitro cultures of bone narrow-derived macrophages were used to determine whether bFGF induced TGF-β1 expression. RESULTS Mice chronically challenged with OVA developed significant airway remodeling that was associated with significantly increased levels of bFGF and TGF-β1. Immunohistochemistry demonstrated significantly increased bFGF and FGF receptor-1 expression by peri- bronchial F4/80+ cells. Double-label immunofluorescence microscopy studies demonstrated that peribronchial macrophages coexpressed bFGF and TGF-β1. In vitro studies demonstrated that incubation of bone marrow-derived macrophages with bFGF induced expression of TGF-β1. Mice treated with corticosteroids and subjected to chronic OVA challenge had significantly reduced levels of bFGF, FGF receptor-1, peribronchial TGF-β1+ cells and airway remodeling. CONCLUSIONS Overall, this study demonstrates that allergen challenge stimulates peribronchial macrophages to coexpress bFGF and TGF-β1 and that bFGF may potentiate macrophage release of TGF-β1 through autocrine and/or paracrine pathways.
Collapse
Affiliation(s)
- Hye Young Yum
- Department of Medicine, University of California San Diego, USA
| | | | | | | |
Collapse
|
13
|
Mohamed JS, Boriek AM. Stretch augments TGF-beta1 expression through RhoA/ROCK1/2, PTK, and PI3K in airway smooth muscle cells. Am J Physiol Lung Cell Mol Physiol 2010; 299:L413-24. [PMID: 20511342 PMCID: PMC2951069 DOI: 10.1152/ajplung.90628.2008] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2008] [Accepted: 05/25/2010] [Indexed: 11/22/2022] Open
Abstract
Transforming growth factor-beta1 (TGF-beta1) expression in smooth muscle cells may play an important role in the pathogenesis of asthma. However, mechanisms that are involved in the regulation of TGF-beta1 gene expression in human airway smooth muscle cells (HASMCs) remain elusive. Here, we show that mechanical stretch of HASMCs augmented TGF-beta1 expression through a de novo RNA synthesis mechanism. Luciferase reporter assays revealed that stretch-induced TGF-beta1 expression was mediated through the enhanced activation of TGF-beta1 promoter. Interestingly, selective inhibitors of PTK, PI3K, or MEK1/2 attenuated TGF-beta1 expression through blocking ERK1/2 phosphorylation and TGF-beta1 promoter activity in response to stretch. In addition, stretch rapidly and transiently augmented GTP-bound RhoA and Rac1 but not Cdc42 GTPase. Either blockade of RhoA GTPase using C3 transferase, ROCK1/2 using Y27632, or knockdown of endogenous RhoA using RhoA siRNA attenuated stretch-induced TGF-beta1 expression through the inhibition of ERK1/2 phosphorylation. Moreover, stretch augmented DNA binding activity of AP-1 in a time-dependent manner. Either treatment of HASMCs with the inhibitors of RhoA, ROCK1/2, PTK, PI3K, MEK1/2, or AP-1 or transfection of HASMCs with AP-1 decoy oligonucleotide attenuated stretch-induced TGF-beta1 expression through repressing the DNA binding activity of AP-1. Site-directed mutagenesis demonstrated that two AP-1 binding sites in the TGF-beta1 promoter region are responsible for stretch-induced TGF-beta1 expression. Overall, in HASMCs, mechanical stretch plays an important role in TGF-beta1 gene upregulation through a stretch-induced signaling pathway, which could be a potential therapeutic intervention for TGF-beta1-induced pathogenesis in asthma.
Collapse
Affiliation(s)
- Junaith Shaik Mohamed
- Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Baylor College of Medicine, Houston, Texas 77030, USA
| | | |
Collapse
|
14
|
Kraniak JM, Sun D, Mattingly RR, Reiners JJ, Tainsky MA. The role of neurofibromin in N-Ras mediated AP-1 regulation in malignant peripheral nerve sheath tumors. Mol Cell Biochem 2010; 344:267-76. [PMID: 20680410 DOI: 10.1007/s11010-010-0551-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2010] [Accepted: 07/23/2010] [Indexed: 11/29/2022]
Abstract
Plexiform neurofibromas commonly found in patients with Neurofibromatosis type I (NF1) have a 5% risk of being transformed into malignant peripheral nerve sheath tumors (MPNST). Germline mutations in the NF1 gene coding for neurofibromin, which is a Ras GTPase activating protein (RasGAP) and a negative regulator of Ras, result in an upregulation of the Ras pathway. We established a direct connection between neurofibromin deficiency and downstream effectors of Ras in cell lines from MPNST patients by demonstrating that knockdown of NF1 expression using siRNA in a NF1 wild type MPNST cell line, STS-26T, activates the Ras/ERK1,2 pathway and increases AP-1 binding and activity. We believe this is the first time the transactivation of AP-1 has been linked directly to neurofibromin deficiency in a disease relevant MPNST cell line. Previously, we have shown that N-Ras is constitutively activated in cell lines derived from independent MPNSTs from NF1 patients. We therefore sought to analyze the role of the N-Ras pathway in deregulating AP-1 transcriptional activity. We show that STS-26T clones conditionally expressing oncogenic N-Ras show increased phosphorylated ERK1,2 and phosphorylated JNK expression concomitant with increased AP-1 activity. MAP kinase pathways (ERK1,2 and JNK) were further examined in ST88-14, a neurofibromin-deficient MPNST cell line. The basal activity of ERK1,2 but not JNK was found to increase AP-1 activity. These experiments further confirmed the link between the loss of neurofibromin and increased activity of Ras/MAP kinase pathways and the activation of downstream transcriptional mechanisms in MPNSTs from NF1 patients.
Collapse
Affiliation(s)
- Janice M Kraniak
- Programs in Molecular Biology and Genetics, Barbara Ann Karmanos Cancer Institute, Detroit, MI 48201, USA
| | | | | | | | | |
Collapse
|
15
|
Sullivan DE, Ferris M, Nguyen H, Abboud E, Brody AR. TNF-alpha induces TGF-beta1 expression in lung fibroblasts at the transcriptional level via AP-1 activation. J Cell Mol Med 2010. [PMID: 20141610 DOI: 10.1111/j.1582-4934.2008.00647.x] [Citation(s) in RCA: 120] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Tumour necrosis factor-alpha (TNF-alpha) and transforming growth factor-beta(1) (TGF-beta(1)) are peptides with multiple biological activities that influence neoplastic, immunologic and fibroproliferative diseases. There are clear interrelationships and overlap between the actions of TNF-alpha and TGF-beta(1) in lung fibrosis; therefore, we postulated that TNF-alpha may play a significant role in regulating TGF-beta(1) expression in lungs. We recently reported that TNF-alpha activates the extracellular regulated kinase (ERK)-specific pathway in fibroblasts resulting in stabilization of TGF-beta(1) mRNA and increased expression of TGF-beta(1). In the current study, we further investigated the molecular mechanisms involved in TNF-alpha regulation of TGF-beta(1) expression. Nuclear run-on assays showed that treatment of Swiss 3T3 fibroblasts with TNF-alpha increased transcription of the TGF-beta(1) gene in an ERK independent manner. Pre-treatment with the activator protein-1 (AP-1) inhibitor curcumin attenuated TNF-alpha induced transcription of the TGF-beta(1) gene. TNF-alpha induced increased levels of c-Jun and C-Fos in the nucleus accompanied by phosphorylation of c-Jun. In electrophoretic mobility shift assays, AP-1 binding to an AP-1 binding site found within the TGF-beta(1) promoter was increased in nuclear extracts from Swiss 3T3 fibroblasts treated with TNF-alpha. Together, these results suggest that TNF-alpha induces expression and DNA binding of AP-1 resulting in increased transcription of the TGF-beta(1) gene. It is essential to know which transcription pathways are activated because of the wide distribution of TNF-alpha and TGF-beta(1), the general lack of effective treatments for fibroproliferative disease and the possibility that targeting the correct transcription factors could be palliative.
Collapse
Affiliation(s)
- Deborah E Sullivan
- Department of Microbiology and Immunology, Biomedical Sciences Graduate Program, Tulane University School of Medicine, New Orleans, LA, USA
| | | | | | | | | |
Collapse
|
16
|
Sullivan DE, Ferris M, Nguyen H, Abboud E, Brody AR. TNF-alpha induces TGF-beta1 expression in lung fibroblasts at the transcriptional level via AP-1 activation. J Cell Mol Med 2010; 13:1866-76. [PMID: 20141610 DOI: 10.1111/j.1582-4934.2009.00647.x] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Tumour necrosis factor-alpha (TNF-alpha) and transforming growth factor-beta(1) (TGF-beta(1)) are peptides with multiple biological activities that influence neoplastic, immunologic and fibroproliferative diseases. There are clear interrelationships and overlap between the actions of TNF-alpha and TGF-beta(1) in lung fibrosis; therefore, we postulated that TNF-alpha may play a significant role in regulating TGF-beta(1) expression in lungs. We recently reported that TNF-alpha activates the extracellular regulated kinase (ERK)-specific pathway in fibroblasts resulting in stabilization of TGF-beta(1) mRNA and increased expression of TGF-beta(1). In the current study, we further investigated the molecular mechanisms involved in TNF-alpha regulation of TGF-beta(1) expression. Nuclear run-on assays showed that treatment of Swiss 3T3 fibroblasts with TNF-alpha increased transcription of the TGF-beta(1) gene in an ERK independent manner. Pre-treatment with the activator protein-1 (AP-1) inhibitor curcumin attenuated TNF-alpha induced transcription of the TGF-beta(1) gene. TNF-alpha induced increased levels of c-Jun and C-Fos in the nucleus accompanied by phosphorylation of c-Jun. In electrophoretic mobility shift assays, AP-1 binding to an AP-1 binding site found within the TGF-beta(1) promoter was increased in nuclear extracts from Swiss 3T3 fibroblasts treated with TNF-alpha. Together, these results suggest that TNF-alpha induces expression and DNA binding of AP-1 resulting in increased transcription of the TGF-beta(1) gene. It is essential to know which transcription pathways are activated because of the wide distribution of TNF-alpha and TGF-beta(1), the general lack of effective treatments for fibroproliferative disease and the possibility that targeting the correct transcription factors could be palliative.
Collapse
Affiliation(s)
- Deborah E Sullivan
- Department of Microbiology and Immunology, Biomedical Sciences Graduate Program, Tulane University School of Medicine, New Orleans, LA, USA
| | | | | | | | | |
Collapse
|
17
|
Lindke AL, Middleton FA, Miller MW. Regulating the availability of transforming growth factor ß1 in B104 neuroblastoma cells. Exp Neurol 2010; 225:123-32. [PMID: 20547156 DOI: 10.1016/j.expneurol.2010.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2010] [Revised: 05/27/2010] [Accepted: 06/07/2010] [Indexed: 10/19/2022]
Abstract
Transforming growth factor (TGF) beta1 is a key player in early brain development, hence, its availability (i.e., synthesis and release) affects neuronogenesis. TGFbeta1 moves proliferating cells out of the cell cycle and promotes their subsequent migration. The present study tested the hypothesis that neural progenitors self-regulate TGFbeta1. B104 neuroblastoma cells which can grow in the absence of serum or growth factors were used in systematic studies of transcription, translation, release, and activation. These studies relied on quantitative enzyme-linked immunosorbent assays and real-time polymerase chain reactions. TGFbeta1 positively upregulated its own intracellular expression and promoted increased release of TGFbeta1 from cells. The induction of TGFbeta1 was independent of a change in transcription, but it depended on cycloheximide-inhibited translation. Signaling mediated by downstream Smad2/3 through the TGFbeta receptors and intracellular protein transport were also required for release of TGFbeta1 from B104 cells. Thus, TGFbeta1 production and release were mediated through a feed-forward mechanism and were pivotally regulated at the level of translation. These activities appear to be key for the role of TGFbeta1 in the proliferation and migration of young neurons.
Collapse
Affiliation(s)
- Amanda L Lindke
- Department of Neuroscience and Physiology, State University of New York-Upstate Medical University, Syracuse NY 13210, USA
| | | | | |
Collapse
|
18
|
Bio-released gold ions modulate expression of neuroprotective and hematopoietic factors after brain injury. Brain Res 2010; 1307:1-13. [DOI: 10.1016/j.brainres.2009.10.028] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2009] [Revised: 10/06/2009] [Accepted: 10/12/2009] [Indexed: 12/21/2022]
|
19
|
Bryant MR, Marta CB, Kim FS, Bansal R. Phosphorylation and lipid raft association of fibroblast growth factor receptor-2 in oligodendrocytes. Glia 2009; 57:935-46. [PMID: 19053057 DOI: 10.1002/glia.20818] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Fibroblast growth factors (FGFs) and their receptors (FGFRs) initiate diverse cellular responses that contribute to the regulation of oligodendrocyte (OL) function. To understand the mechanisms by which FGFRs elicit these cellular responses, we investigated the phosphorylation of signal transduction proteins and the role of cholesterol-glycosphingolipid-enriched "lipid raft" microdomains in differentiated OLs. Surprisingly, we found that the most abundant tyrosine-phosphorylated protein in OLs was the 120-kd isoform of FGFR2 and that it was phosphorylated even in the absence of FGF2, suggesting a potential ligand-independent function for this receptor. Furthermore, FGFR2, but not FGFR1, was associated with lipid raft microdomains in OLs and myelin (but not in astrocytes). This provides the first evidence for the association of FGFR with TX-100-insoluble lipid raft fractions. FGFR2 phosphorylated the key downstream target, FRS2 in OLs. Raft disruption resulted in loss of phosphorylated FRS2 from lipid rafts, coupled with the loss of Akt but not of Mek or Erk phosphorylation. This suggests that FGFR2-FRS2 signaling in lipid rafts operates via the PI3-Kinase/Akt pathway rather than the Ras/Mek/Erk pathway, emphasizing the importance of microenvironments within the cell membrane. Also present in lipid rafts in OLs and myelin, but not in astrocytes, was a novel 52-kd isoform of FGFR2 that lacked the extracellular ligand-binding region. These results demonstrate that FGFR2 in OLs and myelin possess unique characteristics that are specific both to receptor type and to OLs and provide a novel mechanism to elicit distinct cellular responses that mediate both FGF-dependent and -independent functions.
Collapse
Affiliation(s)
- M R Bryant
- Department of Neuroscience, University of Connecticut Medical School, Farmington, Connecticut 06030-3401, USA
| | | | | | | |
Collapse
|
20
|
Pedersen MØ, Jensen R, Pedersen DS, Skjolding AD, Hempel C, Maretty L, Penkowa M. Metallothionein-I+II in neuroprotection. Biofactors 2009; 35:315-25. [PMID: 19655389 DOI: 10.1002/biof.44] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Metallothionein (MT)-I+II synthesis is induced in the central nervous system (CNS) in response to practically any pathogen or disorder, where it is increased mainly in reactive glia. MT-I+II are involved in host defence reactions and neuroprotection during neuropathological conditions, in which MT-I+II decrease inflammation and secondary tissue damage (oxidative stress, neurodegeneration, and apoptosis) and promote post-injury repair and regeneration (angiogenesis, neurogenesis, neuronal sprouting and tissue remodelling). Intracellularly the molecular MT-I+II actions involve metal ion control and scavenging of reactive oxygen species (ROS) leading to cellular redox control. By regulating metal ions, MT-I+II can control metal-containing transcription factors, zinc-finger proteins and p53. However, the neuroprotective functions of MT-I+II also involve an extracellular component. MT-I+II protects the neurons by signal transduction through the low-density lipoprotein family of receptors on the cell surface involving lipoprotein receptor-1 (LRP1) and megalin (LRP2). In this review we discuss the newest data on cerebral MT-I+II functions following brain injury and experimental autoimmune encephalomyelitis.
Collapse
Affiliation(s)
- Mie Ø Pedersen
- Section of Neuroprotection, Institute of Neuroscience and Pharmacology, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | | | | | | |
Collapse
|
21
|
Luckett-Chastain LR, Gallucci RM. Interleukin (IL)-6 modulates transforming growth factor-beta expression in skin and dermal fibroblasts from IL-6-deficient mice. Br J Dermatol 2009; 161:237-48. [PMID: 19438433 DOI: 10.1111/j.1365-2133.2009.09215.x] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Interleukin (IL-6) and transforming growth factor (TGF)-beta have been shown to play a role in skin development and maintenance. OBJECTIVES A link between these two cytokines has yet to be identified and therefore in this study we investigated the modulation of TGF-beta1 and TGF-beta type 2 receptor (TGF-betaR2) by IL-6 in skin. METHODS An IL-6 knockout (IL-6KO) fibroblast-populated lattice model and intradermal injections of IL-6 into unwounded IL-6KO mice were used to investigate the direct effects of IL-6 treatment on TGF-beta and TGF-betaR2 expression and to determine the signalling mechanism. In addition, IL-6KO and C57BL/6 control mice were wounded by a 4-mm punch biopsy to monitor expression of TGF-beta1 and TGF-betaR2 within a wound over time. The expression of TGF-beta1 and TGF-betaR2 was assessed by real-time quantitative polymerase chain reaction, enzyme-linked immunosorbent assay and immunohistology. RESULTS Recombinant IL-6 treatment of IL-6KO lattices and intradermal injections of IL-6 showed a significant induction of TGF-beta1 mRNA and protein, with TGF-beta1 expression localized in the dermis, while TGF-betaR2 expression was primarily in the epidermis in IL-6KO mice. During healing, the expression of TGF-beta1 and TGF-betaR2 mRNA was significantly greater in unwounded and 7-day-old wounds from wild-type mice; however, protein expression did not differ. Treatment with signal transduction inhibitors indicated that IL-6 modulates TGF-beta through a mitogen-activated protein kinase/extracellular signal-regulated kinase (Mapk/Erk)-dependent mechanism. CONCLUSION These studies indicate that IL-6 has the ability to modulate the expression of TGF-beta and TGF-betaR2 to varying degrees in the skin, which may provide a possible mechanism for defining the role of IL-6 in skin maintenance and a new association of IL-6 with TGF-beta in pathologies associated with fibrosis.
Collapse
Affiliation(s)
- L R Luckett-Chastain
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma, Health Sciences Center, Oklahoma City, OK 73117, USA
| | | |
Collapse
|
22
|
Wakade C, King MD, Laird MD, Alleyne CH, Dhandapani KM. Curcumin attenuates vascular inflammation and cerebral vasospasm after subarachnoid hemorrhage in mice. Antioxid Redox Signal 2009; 11:35-45. [PMID: 18752423 DOI: 10.1089/ars.2008.2056] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Cerebral vasospasm is a major cause of death and disability after subarachnoid hemorrhage (SAH); however, clinical therapies to limit the development of cerebral vasospasm are lacking. Although the causative factors underlying the development of cerebral vasospasm are poorly understood, oxidative stress contributes to disease progression. In the present study, curcumin (150 or 300 mg/kg) protected against the development of cerebral vasospasm and limited secondary cerebral infarction after SAH in mice. The protective effect of curcumin was associated with a significant attenuation of inflammatory gene expression and lipid peroxidation within the cerebral cortex and the middle cerebral artery. Despite the ability of curcumin to limit the development of cerebral vasospasm and secondary infarction, behavioral outcome was not improved, indicating a dissociation between cerebral vasospasm and neurologic outcome. Together, these data indicate a novel role for curcumin as a possible adjunct therapy after SAH, both to prevent the development of cerebral vasospasm and to reduce oxidative brain injury after secondary infarction.
Collapse
Affiliation(s)
- Chandramohan Wakade
- Department of Neurosurgery, Medical College of Georgia, Augusta, Georgia 30912, USA
| | | | | | | | | |
Collapse
|
23
|
Laird MD, Wakade C, Alleyne CH, Dhandapani KM. Hemin-induced necroptosis involves glutathione depletion in mouse astrocytes. Free Radic Biol Med 2008; 45:1103-14. [PMID: 18706498 DOI: 10.1016/j.freeradbiomed.2008.07.003] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2008] [Revised: 07/02/2008] [Accepted: 07/07/2008] [Indexed: 01/05/2023]
Abstract
Intracerebral hemorrhage (ICH) is a devastating neurological injury associated with significant mortality. Astrocytic inflammation may contribute to the pathogenesis of ICH, although the underlying cellular mechanisms remain unclear. In this study, the hemoglobin oxidation by-product, hemin, concentration dependently induced necroptotic cell death in cortical astrocytes within 5 h of treatment. Hemin-induced cell death was preceded by increased inflammatory gene expression (COX-2, IL-1beta, TNF-alpha, iNOS). Inhibition of the NF-kappaB transcription factor reversed inflammatory gene expression and attenuated cell death after hemin treatment, suggesting a possible role for inflammatory mediators in astrocytic injury. Superoxide production paralleled the increase in iNOS expression, and inhibition of either iNOS (aminoguanidine or iminopiperdine) or superoxide (apocynin) significantly reduced cell death. Similarly, reduced formation of peroxynitrite, the damaging product of nitric oxide and superoxide, significantly reduced hemin injury. Hemin-induced peroxidative injury was associated with a rapid depletion of intracellular glutathione (GSH), culminating in lipid peroxidation and cell death, effects that were reduced by cotreatment with exogenous GSH, N-acetyl-L-cysteine, or the glutathione peroxidase mimetic ebselen. Together, these studies suggest a novel role for GSH depletion in necroptotic astrocyte injury after a hemorrhagic injury and indicate that therapeutic targeting of GSH may exert a beneficial effect after ICH.
Collapse
Affiliation(s)
- Melissa D Laird
- Department of Neurosurgery, Medical College of Georgia, Augusta, GA 30912, USA
| | | | | | | |
Collapse
|
24
|
Role of TGF-β and FGF in the Treatment of Radiation-Impaired Wounds Using a Novel Drug Delivery System. Plast Reconstr Surg 2008; 122:1036-1045. [DOI: 10.1097/prs.0b013e3181858bf0] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
25
|
Vender JR, Laird MD, Dhandapani KM. Inhibition of NFkappaB reduces cellular viability in GH3 pituitary adenoma cells. Neurosurgery 2008; 62:1122-7; discussion 1027-8. [PMID: 18580810 DOI: 10.1227/01.neu.0000325874.82999.75] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE Adenomas of the pituitary gland are among the most common types of tumors of the adult brain. Although adenomas are histologically benign, they may be associated with significant morbidity and mortality, mostly because of their invasive growth pattern and hormone hypersecretion. Current medical therapies are suppressive, acting at a receptor level. Thus, there is a need to identify novel cellular and molecular targets for pituitary tumors. We investigated the possible role of the NFkappaB transcription factor in pituitary tumor cell growth. METHODS The effect of NFkappaB pathway inhibition on cellular viability was studied in the GH3 pituitary adenoma cell line, a well-characterized rat cell line that secretes growth hormone and prolactin. Cells were treated with mechanistically diverse pharmacological NFkappaB pathway inhibitors or with molecular inhibitors that were overexpressed in tumor cells before the assessment of cellular viability. NFkappaB activity was also assessed in GH3 cells using deoxyribonucleic acid binding assays. RESULTS GH3 cells exhibited constitutive NFkappaB activity, which contributed to increased cellular proliferation. Treatment with wedelolactone, an IkappaB kinase inhibitor, or overexpression of an IkappaB super-repressor reduced cell viability, further implicating NFkappaB in pituitary tumor cell growth. Pharmacological or molecular inhibition of Akt similarly reduced GH3 viability and NFkappaB binding, suggesting that constitutive activation of NFkappaB may be, at least in part, mediated by Akt. CONCLUSION Directed targeting of the Akt and NFkappaB signaling pathways may be a useful adjunct in the clinical management of pituitary tumors. Further elucidation of this pathway may yield novel information regarding the behavior of pituitary tumors in humans.
Collapse
Affiliation(s)
- John R Vender
- Department of Neurosurgery, Medical College of Georgia, Augusta, Georgia 30912, USA
| | | | | |
Collapse
|
26
|
Vender JR, Laird MD, Dhandapani KM. INHIBITION OF NFκB REDUCES CELLULAR VIABILITY IN GH3 PITUITARY ADENOMA CELLS. Neurosurgery 2008. [DOI: 10.1227/01.neu.0000312715.01310.49] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
27
|
BMP signaling through BMPRIA in astrocytes is essential for proper cerebral angiogenesis and formation of the blood-brain-barrier. Mol Cell Neurosci 2008; 38:417-30. [PMID: 18501628 DOI: 10.1016/j.mcn.2008.04.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2007] [Revised: 04/03/2008] [Accepted: 04/04/2008] [Indexed: 11/21/2022] Open
Abstract
Bone morphogenetic protein (BMP) signaling is involved in differentiation of neural precursor cells into astrocytes, but its contribution to angiogenesis is not well characterized. This study examines the role of BMP signaling through BMP type IA receptor (BMPRIA) in early neural development using a conditional knockout mouse model, in which Bmpr1a is selectively disrupted in telencephalic neural stem cells. The conditional mutant mice show a significant increase in the number of cerebral blood vessels and the level of vascular endothelial growth factor (VEGF) is significantly upregulated in the mutant astrocytes. The mutant mice also show leakage of immunoglobulin around cerebral microvessels in neonatal mice, suggesting a defect in formation of the blood-brain-barrier. In addition, astrocytic endfeet fail to encircle cortical blood vessels in the mutant mice. These results suggest that BMPRIA signaling in astrocytes regulates the expression of VEGF for proper cerebrovascular angiogenesis and has a role on in the formation of the blood-brain-barrier.
Collapse
|
28
|
Kim HJ, Ham SA, Kim SU, Hwang JY, Kim JH, Chang KC, Yabe-Nishimura C, Kim JH, Seo HG. Transforming growth factor-beta1 is a molecular target for the peroxisome proliferator-activated receptor delta. Circ Res 2007; 102:193-200. [PMID: 18007025 DOI: 10.1161/circresaha.107.158477] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The peroxisome proliferator-activated receptor (PPAR)delta has been implicated in the pathogenesis of atherogenic disorders. However, its physiological roles and functions in vascular smooth muscle cells (VSMCs) remain relatively unclear. In the present study, we show that the gene encoding transforming growth factor (TGF)-beta1 is a PPARdelta target in VSMCs. The PPARdelta activator GW501516 upregulates TGF-beta1 expression in a dose- and time-dependent manner. This induction is attenuated significantly by the presence of small interfering RNA against PPARdelta or GW9662, an inhibitor of PPARdelta. Furthermore, activated PPARdelta induces TGF-beta1 promoter activity by binding to the direct repeat-1 response element TGF-beta1-direct repeat-1. Mutations in the 5' or 3' half-sites of the response element totally abrogate transcriptional activation and PPARdelta binding, which suggests that this site is a novel type of PPARdelta response element. In addition, ligand-activated PPARdelta attenuated the promoter activity and expression of monocyte chemoattractant protein-1 induced by interleukin-1beta. These effects were significantly reduced in the presence of small interfering RNA against PPARdelta, anti-TGF-beta1 antibody, or a TGF-beta type I receptor inhibitor. Decreased monocyte chemoattractant protein-1 expression induced by PPARdelta was mediated by the effector of TGF-beta1, Smad3. Finally, administration of GW501516 to mice upregulated TGF-beta1, whereas the expression of proinflammatory genes including monocyte chemoattractant protein-1 was significantly attenuated in the thoracic aorta. Taken together, these results demonstrate the presence of a novel TGF-beta1-mediated pathway in the antiinflammatory activities of PPARdelta.
Collapse
Affiliation(s)
- Hyo Jung Kim
- Department of Pharmacology, Gyeongsang Institute of Health Science, Gyeongsang National University School of Medicine, Jinju, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Bülow R, Fitzner B, Sparmann G, Emmrich J, Liebe S, Jaster R. Antifibrogenic effects of histone deacetylase inhibitors on pancreatic stellate cells. Biochem Pharmacol 2007; 74:1747-57. [PMID: 17889833 DOI: 10.1016/j.bcp.2007.08.023] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2007] [Revised: 08/16/2007] [Accepted: 08/16/2007] [Indexed: 12/11/2022]
Abstract
Pancreatic stellate cells (PSCs) are essentially involved in pancreatic fibrogenesis and considered as a target for antifibrotic therapies. Here, we have analyzed the effects of three histone deacetylase inhibitors (HDACIs), sodium butyrate, sodium valproate (VPA) and trichostatin A (TSA), on profibrogenic activities of PSC and elucidated molecular targets of HDACI action. Therefore, cultured PSCs were exposed to HDACI. Cell proliferation and viability were assessed by 5-bromo-2'-deoxyuridine (BrdU) incorporation and trypan blue staining assays. Exhibition of the myofibroblastic PSC phenotype was monitored by immunofluorescence analysis of alpha-smooth muscle actin (alpha-SMA) expression. [(3)H]-proline incorporation into acetic acid-soluble proteins was measured to quantify collagen synthesis. Levels of mRNA were determined by quantitative reverse transcriptase real-time PCR. Protein expression, phosphorylation and acetylation were analyzed by immunoblotting, and gel shift assays were performed to study DNA binding of nuclear proteins. HDACI enhanced histone H3 acetylation in a dose-dependent manner. In the same dose range, they strongly inhibited cell proliferation, alpha-SMA expression and collagen synthesis. A significantly increased rate of cell death was observed in response to TSA at 1 microM. While all three HDACI inhibited mRNA expression of endothelin-1, only VPA significantly reduced expression of transforming growth factor-beta1. Both mediators exert autocrine profibrogenic effects on PSC. Furthermore, HDACI-treated PSC displayed a diminished DNA binding of AP-1, a key transcription factor in profibrogenic signaling. Together, the results suggest that HDACI exert antifibrogenic effects on PSC. Interruption of AP-1 signaling and autocrine loops enhancing PSC activation might be key mechanisms of HDACI action.
Collapse
Affiliation(s)
- Robin Bülow
- Department of Medicine, Division of Gastroenterology, Medical Faculty, University of Rostock, E.-Heydemann-Str. 6, 18057 Rostock, Germany
| | | | | | | | | | | |
Collapse
|
30
|
Zhang Y, Ye LP, Wang B, Cao SC, Sun LG. Effect of lead on ERK activity and the protective function of bFGF in rat primary culture astroglia. J Zhejiang Univ Sci B 2007; 8:422-7. [PMID: 17565513 PMCID: PMC1879161 DOI: 10.1631/jzus.2007.b0422] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
OBJECTIVE To observe the effects of lead on levels of phosphorylated extracellular signal regulated kinase (p-ERK) in the cytoplasm of primary cultures of rat astroglial cells and the possible protective effect of basic fibroblast growth factor (bFGF) on lead-induced effects. METHODS The primary astroglia cells from 1~6 d old Wistar rats were cultured. The cells pretreated with the MEK1 (mitogen-activated protein kinase kinase 1) inhibitor PD98059 and bFGF, respectively, were exposed to Pb acetate of different concentrations for different times. Western blotting and reverse transcription polymerase chain reaction (RT-PCR) methods were used to detect the protein and mRNA expressions of ERK. RESULTS mRNA expression for ERK peaked 15 min after initiation of lead exposure (P<0.05) and protein expression of p-ERK peaked at 30 min (P<0.05). ERK mRNA levels and p-ERK protein levels returned to baseline after 60 and 120 min of lead exposure, respectively (P>0.05). The increase in p-ERK levels in lead-treated cells could be inhibited by PD098059. Activation of ERK in the cells by lead was prevented by pretreatment with bFGF. Total ERK protein levels did not change under the same experimental conditions (P>0.05). CONCLUSION Low-level lead exposure resulted in transient activation of ERK through the MEK pathway, which then returned to basal levels in the continued presence of lead. Exogenous bFGF protected ERK signaling components in astroglia from lead poisoning.
Collapse
|