451
|
Cao J, Feng J, Li L, Chen B. Obstructive sleep apnea promotes cancer development and progression: a concise review. Sleep Breath 2015; 19:453-7. [PMID: 25643765 DOI: 10.1007/s11325-015-1126-x] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Revised: 12/16/2014] [Accepted: 12/23/2014] [Indexed: 12/17/2022]
|
452
|
Wang L, Chen G, Lu X, Wang S, Han S, Li Y, Ping G, Jiang X, Li H, Yang J, Wu C. Novel chalcone derivatives as hypoxia-inducible factor (HIF)-1 inhibitor: Synthesis, anti-invasive and anti-angiogenic properties. Eur J Med Chem 2015; 89:88-97. [PMID: 25462229 DOI: 10.1016/j.ejmech.2014.10.036] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Revised: 09/16/2014] [Accepted: 10/12/2014] [Indexed: 12/17/2022]
|
453
|
Li J, Ke Y, Huang M, Huang S, Liang Y. Inhibitory effects of B-cell lymphoma 2 on the vasculogenic mimicry of hypoxic human glioma cells. Exp Ther Med 2014; 9:977-981. [PMID: 25667663 PMCID: PMC4316972 DOI: 10.3892/etm.2014.2162] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Accepted: 10/29/2014] [Indexed: 12/13/2022] Open
Abstract
The aim of this study was to investigate the mechanisms and effects of B-cell lymphoma 2 (Bcl-2) on the vasculogenic mimicry (VM) of human glioma cells. U87 cells were cultured under hypoxic conditions and then divided into four groups: Control, 3-(5-hydroxymethyl-2-furyl)-1-benzylindazole (YC-1), ABT-737 and YC-1 + ABT-737. These groups were treated with the corresponding simulators. The expression of hypoxia-inducible factor-1α (HIF-1α), matrix metalloproteinase (MMP)-2, MMP-14 and Bcl-2 in each group was determined using a reverse transcription-quantitative polymerase chain reaction and western blot analysis. Compared with that in the control group, the mRNA and protein expression of MMP-2, MMP-14 and Bcl-2 in the YC-1 and ABT-737 groups was significantly reduced. The expression of HIF-1α, however, was only significantly reduced in the YC-1 group (P<0.05). Compared with those in the YC-1 + ABT-737 group, the expression levels of the four proteins in the YC-1 and ABT-737 groups were not significantly different, with the exception of the expression of HIF-1α in the ABT-737 group, which was significantly enhanced (P<0.05). The mRNA expression levels of HIF-1α, MMP-2 and MMP-14 in the YC-1 group were significantly different from those in the ABT-737 group (P<0.01); however, no significant difference was observed in the expression of Bcl-2. In conclusion, Bcl-2 may be an important factor in the VM formation of human malignant glioma U87 cells under hypoxic conditions. Certain functions of Bcl-2 may be attributed to the HIF-1α-MMP-2-MMP-14-VM channel, whereas other functions may be independent of the channel.
Collapse
Affiliation(s)
- Jianwen Li
- Department of Neurosurgery, Neurosurgery Institute, Key Laboratory on Brain Function Repair and Regeneration of Guangdong, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong 510282, P.R. China
| | - Yiquan Ke
- Department of Neurosurgery, Neurosurgery Institute, Key Laboratory on Brain Function Repair and Regeneration of Guangdong, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong 510282, P.R. China
| | - Min Huang
- Department of Neurosurgery, Neurosurgery Institute, Key Laboratory on Brain Function Repair and Regeneration of Guangdong, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong 510282, P.R. China
| | - Shuyun Huang
- Department of Neurosurgery, Neurosurgery Institute, Key Laboratory on Brain Function Repair and Regeneration of Guangdong, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong 510282, P.R. China
| | - Yiming Liang
- Department of Neurosurgery, Neurosurgery Institute, Key Laboratory on Brain Function Repair and Regeneration of Guangdong, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong 510282, P.R. China
| |
Collapse
|
454
|
Aaltonen V, Kinnunen K, Jouhilahti EM, Peltonen J, Nikinmaa M, Kaarniranta K, Arjamaa O. Hypoxic conditions stimulate the release of B-type natriuretic peptide from human retinal pigment epithelium cell culture. Acta Ophthalmol 2014; 92:740-4. [PMID: 24739438 DOI: 10.1111/aos.12415] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Accepted: 03/09/2014] [Indexed: 11/30/2022]
Abstract
PURPOSE A-type peptide, a natriuretic peptide belonging to the natriuretic peptide family, has been shown to be increased in the vitreous of patients suffering from diabetic retinopathy and that human retina has a well-developed natriuretic peptide system. The stimulus to which the synthesis of natriuretic peptides responded in these patients has, however, remained unknown. As the natriuretic peptides have recently been shown to respond to hypoxic conditions, the genes of both A-type and B-type have a hypoxia-response element (HRE) in their promoter sequence, we therefore hypothesized that hypoxia in the human retinal pigment epithelium will increase the secretion of NT-proBNP, the most common natriuretic peptide monitored in clinical medicine. METHODS We used cultured human retinal pigment epithelium cell line (ARPE-19) which was exposed either to normoxia or to hypoxia for 2 hr, 4 hr, 6 hr and 24 hr. NT-proBNP was measured with enzyme immunoassay, VEGF with ELISA and HIF-1α with Western blotting. RESULTS Hypoxia induced VEGF 165 release in culture medium and HIF-1α expression in cultured ARPE-19 cells. Time-dependent NT-proBNP release was detected when the ARPE-19 cells were cultured under normoxia. When hypoxia was induced, a statistically significant increase in NT-proBNP release was demonstrated in the culture medium. CONCLUSIONS Hypoxic conditions increase the release of a natriuretic peptide from retinal pigment epithelium (RPE) cells. The secretion of VEGF was also enhanced. The responses were associated with the up-regulation of the HIF-1α transcription factor. These results explain the previous findings from patients with diabetes, which also suggest that hypoxia is a ubiquitous stimulus for the secretion of natriuretic peptides in human body.
Collapse
Affiliation(s)
- Vesa Aaltonen
- Department of Ophthalmology; University Central Hospital; University of Turku; Turku Finland
- Department of Cell Biology and Anatomy; University of Turku; Turku Finland
| | - Kati Kinnunen
- Department of Ophthalmology; University of Eastern Finland; Kuopio Finland
| | | | - Juha Peltonen
- Department of Cell Biology and Anatomy; University of Turku; Turku Finland
| | - Mikko Nikinmaa
- Department of Biology; University of Turku; Turku Finland
| | - Kai Kaarniranta
- Department of Ophthalmology; University of Eastern Finland; Kuopio Finland
| | - Olli Arjamaa
- Department of Biology; University of Turku; Turku Finland
| |
Collapse
|
455
|
Hypoxia-inducible factor 1-α in chronic gastrointestinal ischemia. Virchows Arch 2014; 466:125-32. [PMID: 25425478 DOI: 10.1007/s00428-014-1691-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Revised: 09/29/2014] [Accepted: 11/13/2014] [Indexed: 01/23/2023]
|
456
|
Liang J, Zhang Z, Liang L, Shen Y, Ouyang K. HIF-1α regulated tongue squamous cell carcinoma cell growth via regulating VEGF expression in a xenograft model. ANNALS OF TRANSLATIONAL MEDICINE 2014; 2:92. [PMID: 25405167 DOI: 10.3978/j.issn.2305-5839.2014.08.01] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Accepted: 07/21/2014] [Indexed: 11/14/2022]
Abstract
OBJECTIVE We aimed to study the mechanism of hypoxia-inducible factor 1 alpha (HIF-1α) regulating the cell proliferation of tongue squamous cell carcinoma (TSCC) via vascular endothelial growth factor (VEGF). METHODS We used RNA interference (RNAi) technique, transfected chemically synthesized siRNA against HIF-1α into CAL-27 cells, and detected the expression of HIF-1α and VEGF by real time-PCR and Western blotting in order to find out if HIF-1α regulated the expression of VEGF. A xenograft experiment was carried out to observe the role of HIF-1α on the tumor growth of tongue squamous cell carcinoma. RESULTS HIF-1α and VEGF mRNA expression was significantly downregulated 36 and 48 h after transfection (P<0.05); the protein expression of HIF-1α and VEGF was also significantly suppressed by siRNA against HIF-1α. Furthermore, intratumoraly injection of HIF-1α targeting siRNA suppressed tumor growth in nude mice. CONCLUSIONS HIF-1α regulated VEGF expression, and they may contribute to TSCC cell tumor growth.
Collapse
Affiliation(s)
- Jun Liang
- 1 Department of Oral and Maxillofacial Surgery, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China ; 2 Department of Stomatology, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510655, China ; 3 Department of Stomatology, Shanghai Tenth People's Hospital, Shanghai 200072, China ; 4 Department of Oral and Maxillofacial Surgery, Stomatological Hospital of Guangzhou Medical College, Guangzhou 510140, China
| | - Zhaoqiang Zhang
- 1 Department of Oral and Maxillofacial Surgery, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China ; 2 Department of Stomatology, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510655, China ; 3 Department of Stomatology, Shanghai Tenth People's Hospital, Shanghai 200072, China ; 4 Department of Oral and Maxillofacial Surgery, Stomatological Hospital of Guangzhou Medical College, Guangzhou 510140, China
| | - Lizhong Liang
- 1 Department of Oral and Maxillofacial Surgery, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China ; 2 Department of Stomatology, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510655, China ; 3 Department of Stomatology, Shanghai Tenth People's Hospital, Shanghai 200072, China ; 4 Department of Oral and Maxillofacial Surgery, Stomatological Hospital of Guangzhou Medical College, Guangzhou 510140, China
| | - Yun Shen
- 1 Department of Oral and Maxillofacial Surgery, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China ; 2 Department of Stomatology, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510655, China ; 3 Department of Stomatology, Shanghai Tenth People's Hospital, Shanghai 200072, China ; 4 Department of Oral and Maxillofacial Surgery, Stomatological Hospital of Guangzhou Medical College, Guangzhou 510140, China
| | - Kexiong Ouyang
- 1 Department of Oral and Maxillofacial Surgery, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, China ; 2 Department of Stomatology, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510655, China ; 3 Department of Stomatology, Shanghai Tenth People's Hospital, Shanghai 200072, China ; 4 Department of Oral and Maxillofacial Surgery, Stomatological Hospital of Guangzhou Medical College, Guangzhou 510140, China
| |
Collapse
|
457
|
Wang L, Bhatta A, Toque HA, Rojas M, Yao L, Xu Z, Patel C, Caldwell RB, Caldwell RW. Arginase inhibition enhances angiogenesis in endothelial cells exposed to hypoxia. Microvasc Res 2014; 98:1-8. [PMID: 25445030 DOI: 10.1016/j.mvr.2014.11.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Revised: 10/29/2014] [Accepted: 11/02/2014] [Indexed: 10/24/2022]
Abstract
Hypoxia-induced arginase elevation plays an essential role in several vascular diseases but influence of arginase on hypoxia-mediated angiogenesis is completely unknown. In this study, in vitro network formation in bovine aortic endothelial cells (BAEC) was examined after exposure to hypoxia for 24h with or without arginase inhibition. Arginase activity, protein levels of the two arginase isoforms, eNOS, and VEGF as well as production of NO and ROS were examined to determine the involvement of arginase in hypoxia-mediated angiogenesis. Hypoxia elevated arginase activity and arginase 2 expression but reduced active p-eNOS(Ser1177) and NO levels in BAEC. In addition, both VEGF protein levels and endothelial elongation and network formation were reduced with continued hypoxia, whereas ROS levels increased and NO levels decreased. Arginase inhibition limited ROS, restored NO formation and VEGF expression, and prevented the reduction of angiogenesis. These results suggest a fundamental role of arginase activity in regulating angiogenic function.
Collapse
Affiliation(s)
- Lin Wang
- Department of Plastic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, PR China; Department of Pharmacology and Toxicology, Georgia Regents University, Augusta, GA 30912, USA
| | - Anil Bhatta
- Department of Pharmacology and Toxicology, Georgia Regents University, Augusta, GA 30912, USA
| | - Haroldo A Toque
- Department of Pharmacology and Toxicology, Georgia Regents University, Augusta, GA 30912, USA
| | - Modesto Rojas
- Vascular Biology Center, Georgia Regents University, Charlie Norwood VA Medical Center, Augusta GA, 30912, USA
| | - Lin Yao
- Department of Pharmacology and Toxicology, Georgia Regents University, Augusta, GA 30912, USA
| | - Zhimin Xu
- Vascular Biology Center, Georgia Regents University, Charlie Norwood VA Medical Center, Augusta GA, 30912, USA
| | - Chintan Patel
- Vascular Biology Center, Georgia Regents University, Charlie Norwood VA Medical Center, Augusta GA, 30912, USA
| | - Ruth B Caldwell
- Vascular Biology Center, Georgia Regents University, Charlie Norwood VA Medical Center, Augusta GA, 30912, USA
| | - R William Caldwell
- Department of Pharmacology and Toxicology, Georgia Regents University, Augusta, GA 30912, USA.
| |
Collapse
|
458
|
Koide N, Odkhuu E, Naiki Y, Tsolmongyn B, Ito K, Komatsu T, Yoshida T, Yokochi T. Augmentation of LPS-induced vascular endothelial cell growth factor production in macrophages by transforming growth factor-β1. Innate Immun 2014; 20:816-825. [PMID: 24225655 DOI: 10.1177/1753425913509291] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2025] Open
Abstract
The effect of LPS on the production of vascular endothelial growth factor (VEGF) was examined using RAW 264.7 macrophage cells. LPS induced VEGF production in RAW 264.7 cells and mouse peritoneal cells. LPS induced VEGF production via the expression of hypoxia inducible factor-1α and LPS-induced VEGF production was dependent on the activation of p38 MAPK and NF-κB activation· Transforming growth factor (TGF)-β1 augmented LPS-induced VEGF production, although TGF-β1 alone did not induce VEGF production. The augmentation of LPS-induced VEGF production by TGF-β1 was inhibited by a p38 MAPK inhibitor and was correlated with the phosphorylation of Smad3. The enhancing effect of TGF-β1 on LPS-induced VEGF production was observed in vivo in the skin lesions of mice receiving a subcutaneous injection of LPS. Taken together, it is suggested that LPS induced the VEGF production in macrophages and that it was augmented by TGF-β1 in vitro and in vivo.
Collapse
Affiliation(s)
- Naoki Koide
- Department of Microbiology and Immunology, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Erdenezaya Odkhuu
- Department of Microbiology and Immunology, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Yoshikazu Naiki
- Department of Microbiology and Immunology, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Bilegtsaikhan Tsolmongyn
- Department of Microbiology and Immunology, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Kiyoaki Ito
- Department of Microbiology and Immunology, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Takayuki Komatsu
- Department of Microbiology and Immunology, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Tomoaki Yoshida
- Department of Microbiology and Immunology, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Takashi Yokochi
- Department of Microbiology and Immunology, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| |
Collapse
|
459
|
Lin CY, Hung SY, Chen HT, Tsou HK, Fong YC, Wang SW, Tang CH. Brain-derived neurotrophic factor increases vascular endothelial growth factor expression and enhances angiogenesis in human chondrosarcoma cells. Biochem Pharmacol 2014; 91:522-533. [PMID: 25150213 DOI: 10.1016/j.bcp.2014.08.008] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Revised: 08/11/2014] [Accepted: 08/11/2014] [Indexed: 01/07/2023]
Abstract
Chondrosarcomas are a type of primary malignant bone cancer, with a potent capacity for local invasion and distant metastasis. Brain-derived neurotrophic factor (BDNF) is commonly upregulated during neurogenesis. The aim of the present study was to examine the mechanism involved in BDNF-mediated vascular endothelial growth factor (VEGF) expression and angiogenesis in human chondrosarcoma cells. Here, we knocked down BDNF expression in chondrosarcoma cells and assessed their capacity to control VEGF expression and angiogenesis in vitro and in vivo. We found knockdown of BDNF decreased VEGF expression and abolished chondrosarcoma conditional medium-mediated angiogenesis in vitro as well as angiogenesis effects in vivo in the chick chorioallantoic membrane and Matrigel plug nude mouse models. In addition, in the xenograft tumor angiogenesis model, the knockdown of BDNF significantly reduced tumor growth and tumor-associated angiogenesis. BDNF increased VEGF expression and angiogenesis through the TrkB receptor, PLCγ, PKCα, and the HIF-1α signaling pathway. Finally, we analyzed samples from chondrosarcoma patients by immunohistochemical staining. The expression of BDNF and VEGF protein in 56 chondrosarcoma patients was significantly higher than in normal cartilage. In addition, the high level of BDNF expression correlated strongly with VEGF expression and tumor stage. Taken together, our results indicate that BDNF increases VEGF expression and enhances angiogenesis through a signal transduction pathway that involves the TrkB receptor, PLCγ, PKCα, and the HIF-1α. Therefore, BDNF may represent a novel target for anti-angiogenic therapy for human chondrosarcoma.
Collapse
Affiliation(s)
- Chih-Yang Lin
- Graduate Institute of Basic Medical Science, China Medical University, No. 91, Hsueh-Shih Road, Taichung, Taiwan
| | - Shih-Ya Hung
- Epigenome Research Center, China Medical University Hospital, Taichung, Taiwan; Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Hsien-Te Chen
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan; Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan; Department of Materials Science and Engineering, Feng Chia University, Taichung, Taiwan
| | - Hsi-Kai Tsou
- Department of Materials Science and Engineering, Feng Chia University, Taichung, Taiwan; Department of Neurosurgery, Taichung Veterans General Hospital, Taichung, Taiwan; Department of Early Childhood Care and Education, Jen-Teh Junior College of Medicine, Nursing and Management, Miaoli County, Taiwan
| | - Yi-Chin Fong
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan; Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan
| | - Shih-Wei Wang
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan
| | - Chih-Hsin Tang
- Graduate Institute of Basic Medical Science, China Medical University, No. 91, Hsueh-Shih Road, Taichung, Taiwan; Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan; Department of Biotechnology, College of Health Science, Asia University, Taichung, Taiwan.
| |
Collapse
|
460
|
Madrigal M, Rao KS, Riordan NH. A review of therapeutic effects of mesenchymal stem cell secretions and induction of secretory modification by different culture methods. J Transl Med 2014; 12:260. [PMID: 25304688 PMCID: PMC4197270 DOI: 10.1186/s12967-014-0260-8] [Citation(s) in RCA: 434] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2014] [Accepted: 09/10/2014] [Indexed: 02/06/2023] Open
Abstract
The mesenchymal stem cell (MSC) is being broadly studied in clinical trials. Contrary to the early paradigm of cell replacement and differentiation as a therapeutic mechanism of action, evidence is mounting that the secretions of the cells are responsible for their therapeutic effects. These secretions include molecules and extracellular vesicles that have both local and distant effects. This review summarizes the up- and down-regulation of MSC anti-inflammatory, immune modulating, anti-tumor, and regenerative secretions resulting from different stimuli including: a) hypoxia, which increases the production of growth factors and anti-inflammatory molecules; b) pro-inflammatory stimuli that induce the secretion of immune modulating and anti-inflammatory factors; and c) 3 dimensional growth which up regulates the production of anti-cancer factors and anti-inflammatory molecules compared to monolayer culture. Finally we review in detail the most important factors present in conditioned medium of MSC that can be considered protagonists of MSC physiological effects including HGF, TGF-b, VEGF, TSG-6, PGE2 and galectins 1, and 9. We conclude that there is potential for the development of acellular therapeutic interventions for autoimmune, inflammatory, and malignant diseases and tissue regeneration from cellular secretions derived from MSCs cultured under the appropriate conditions.
Collapse
Affiliation(s)
- Marialaura Madrigal
- Department of Biotechnology, Acharya Nagarjuna University, Guntur, India. .,INDICASAT-AIP, City of Knowledge, Republic of Panama. .,MediStem Panama Inc., City of Knowledge, Republic of Panama.
| | | | - Neil H Riordan
- MediStem Panama Inc., City of Knowledge, Republic of Panama.
| |
Collapse
|
461
|
Brzozowski T. Role of renin-angiotensin system and metabolites of angiotensin in the mechanism of gastric mucosal protection. Curr Opin Pharmacol 2014; 19:90-8. [PMID: 25238456 DOI: 10.1016/j.coph.2014.08.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 08/29/2014] [Accepted: 08/29/2014] [Indexed: 12/29/2022]
Abstract
Angiotensin II, the main effector of the renin-angiotensin system (RAS), is generated from the precursor angiotensinogen by the actions of renin, angiotensin-converting enzyme, chymase and various peptidases. RAS is essential in the control of blood pressure and body fluid homeostasis but their involvement in the mechanism of the protection of gastric mucosa has not been extensively studied. On the other hand, angiotensin-(1-7) which acts on the Mas receptor, exhibits a potent vasodilatory activity and attenuates the gastric lesions induced by various ulcerogens. In this review, the mechanism of RAS, the antagonists of angiotensin AT1 and AT2 receptors and angiotensin-(1-7) in formation of gastric damage is discussed with possible translating relevance to treatment modalities in the protection against gastric mucosal injury.
Collapse
Affiliation(s)
- Tomasz Brzozowski
- Department of Physiology, Jagiellonian University Medical College, Cracow, Poland.
| |
Collapse
|
462
|
Ruthenborg RJ, Ban JJ, Wazir A, Takeda N, Kim JW. Regulation of wound healing and fibrosis by hypoxia and hypoxia-inducible factor-1. Mol Cells 2014; 37:637-43. [PMID: 24957212 PMCID: PMC4179131 DOI: 10.14348/molcells.2014.0150] [Citation(s) in RCA: 141] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Accepted: 06/08/2014] [Indexed: 12/12/2022] Open
Abstract
Wound healing is a complex multi-step process that requires spatial and temporal orchestration of cellular and non-cellular components. Hypoxia is one of the prominent microenvironmental factors in tissue injury and wound healing. Hypoxic responses, mainly mediated by a master transcription factor of oxygen homeostasis, hypoxia-inducible factor-1 (HIF-1), have been shown to be critically involved in virtually all processes of wound healing and remodeling. Yet, mechanisms underlying hypoxic regulation of wound healing are still poorly understood. Better understanding of how the wound healing process is regulated by the hypoxic microenvironment and HIF-1 signaling pathway will provide insight into the development of a novel therapeutic strategy for impaired wound healing conditions such as diabetic wound and fibrosis. In this review, we will discuss recent studies illuminating the roles of HIF-1 in physiologic and pathologic wound repair and further, the therapeutic potentials of HIF-1 stabilization or inhibition.
Collapse
Affiliation(s)
- Robin J Ruthenborg
- Department of Molecular and Cell Biology, The University of Texas at Dallas, Richardson TX 75080, USA
| | - Jae-Jun Ban
- Department of Molecular and Cell Biology, The University of Texas at Dallas, Richardson TX 75080, USA
| | - Anum Wazir
- Department of Molecular and Cell Biology, The University of Texas at Dallas, Richardson TX 75080, USA
| | | | - Jung-whan Kim
- Department of Molecular and Cell Biology, The University of Texas at Dallas, Richardson TX 75080, USA
| |
Collapse
|
463
|
Andreeva K, Zhang M, Fan W, Li X, Chen Y, Rebolledo-Mendez JD, Cooper NG. Time-dependent Gene Profiling Indicates the Presence of Different Phases for Ischemia/Reperfusion Injury in Retina. OPHTHALMOLOGY AND EYE DISEASES 2014; 6:43-54. [PMID: 25210480 PMCID: PMC4149383 DOI: 10.4137/oed.s17671] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Revised: 07/09/2014] [Accepted: 07/10/2014] [Indexed: 02/05/2023]
Abstract
Ischemia/reperfusion (IR) injury has been associated with several retinal pathologies, and a few genes/gene products have been linked to IR injury. However, the big picture of temporal changes, regarding the affected gene networks, pathways, and processes remains to be determined. The purpose of the present study was to investigate initial, intermediate, and later stages to characterize the etiology of IR injury in terms of the pathways affected over time. Analyses indicated that at the initial stage, 0-hour reperfusion following the ischemic period, the ischemia-associated genes were related to changes in metabolism. In contrast, at the 24-hour time point, the signature events in reperfusion injury include enhanced inflammatory and immune responses as well as cell death indicating that this would be a critical period for the development of any interventional therapeutic strategies. Genes in the signal transduction pathways, particularly transmitter receptors, are downregulated at this time. Activation of the complement system pathway clearly plays an important role in the later stages of reperfusion injury. Together, these results demonstrate that the etiology of injury related to IR is characterized by the appearance of specific patterns of gene expression at any given time point during retinal IR injury. These results indicate that evaluation of treatment strategies with respect to time is very critical.
Collapse
Affiliation(s)
- Kalina Andreeva
- Department of Anatomical Sciences and Neurobiology, University of Louisville, School of Medicine, Louisville, KY, USA
| | - Meixia Zhang
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Wei Fan
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiaohong Li
- Department of Anatomical Sciences and Neurobiology, University of Louisville, School of Medicine, Louisville, KY, USA
| | - Yinlu Chen
- Department of Anatomical Sciences and Neurobiology, University of Louisville, School of Medicine, Louisville, KY, USA
| | - Jovan D Rebolledo-Mendez
- Department of Anatomical Sciences and Neurobiology, University of Louisville, School of Medicine, Louisville, KY, USA
| | - Nigel G Cooper
- Department of Anatomical Sciences and Neurobiology, University of Louisville, School of Medicine, Louisville, KY, USA
| |
Collapse
|
464
|
Darakhshan S, Bidmeshkipour A, Khazaei M, Rabzia A, Ghanbari A. Synergistic effects of tamoxifen and tranilast on VEGF and MMP-9 regulation in cultured human breast cancer cells. Asian Pac J Cancer Prev 2014; 14:6869-74. [PMID: 24377619 DOI: 10.7314/apjcp.2013.14.11.6869] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Vascular endothelial growth factor and matrix metalloproteinases are two important factors for angiogenesis associated with breast cancer growth and progression. The present study was aimed to examine the effects of tamoxifen and tranilast drugs singly or in combination on proliferation of breast cancer cells and also to evaluate VEGF and MMP-9 expression and VEGF secretion levels. MATERIALS AND METHODS Human breast cancer cell lines, MCF-7 and MDA-MB-231, were treated with tamoxifen and/or tranilast alone or in combination and percentage cell survival and proliferative activity were evaluated using LDH leakage and MTT assays. mRNA expression and protein levels were examined by real-time RT-PCR and ELISA assay, respectively. RESULTS LDH and MTT assays showed that the combined treatment of tamoxifen and tranilast resulted in a significant decrease in cell viability and cell proliferation compared with tamoxifen or tranilast treatment alone, with significant decrease in VEGF mRNA and protein levels. We also found that tamoxifen as a single agent rarely increased MMP-9 expression. A decrease in MMP-9 expression was seen after treatment with tranilast alone and in the combined treatment MMP-9 mRNA level was decreased. CONCLUSIONS This combination treatment can able to inhibit growth, proliferation and angiogenesis of breast cancer cells.
Collapse
Affiliation(s)
- Sara Darakhshan
- Department of Biology, Faculty of Science, Razi University, Kermanshah, Iran E-mail :
| | | | | | | | | |
Collapse
|
465
|
Healed porcine incisions previously treated with a surgical incision management system: mechanical, histomorphometric, and gene expression properties. Aesthetic Plast Surg 2014; 38:767-78. [PMID: 24912426 DOI: 10.1007/s00266-014-0339-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2013] [Accepted: 04/18/2014] [Indexed: 01/14/2023]
Abstract
BACKGROUND Computer and bench models have shown previously that surgical incision management with negative pressure (SIM) immediately decreases lateral tissue tension and increases incisional apposition. Better apposition is known to improve healing. Thus, SIM was hypothesized to improve the quality of incisional healing. This study evaluated the impact that 5 days of SIM had on mechanical properties and associated changes in the histology/histomorphometry and gene expression of healed porcine incisions. METHODS One incision in each of the 4 pairs of contralateral, sutured, full-thickness incisions in each of 6 Yucatan swine were treated with either SIM (Prevena™ Incision Management System; n = 24 incisions/treatment group) or standard of care (SOC; sterile absorbent abdominal pads; n = 24/group) for 5 days, after which both groups received SOC for an additional 5 days. Biopsies for gene-expression analyses were collected on days 5 (n = 6 pairs/group), 20 (n = 6 pairs/group), and 40 (n = 12 pairs/group). On day 40, the animals were killed, after which healed incisions were harvested for mechanical testing (n = 12/group) and histologic/histomorphometric evaluation (n = 12/group). RESULTS Compared with SOC-treated incisions, SIM-treated incisions had significantly improved (p < 0.05) mechanical properties (strain energy density, peak strain) and a narrower scar/healed area in the deep dermis on day 40. Differences in gene expression between SOC- and SIM-treated specimens were observed primarily on day 5. The SIM-treated specimens had significantly fewer genes, which were differentially expressed and showed reduced upregulation of genes associated with inflammation, hypoxia, retardation of reepithelialization, impaired wound healing, and scarring. CONCLUSION Early application of SIM improved the quality of healed porcine incisions in terms of mechanical, histomorphometric, and gene-expression properties. NO LEVEL ASSIGNED This journal requires that authors assign a level of evidence to each submission to which Evidence-Based Medicine rankings are applicable. This excludes Review Articles, Book Reviews, and manuscripts that concern Basic Science, Animal Studies, Cadaver Studies, and Experimental Studies. For a full description of these Evidence-Based Medicine ratings, please refer to the Table of Contents or the online Instructions to Authors www.springer.com/00266 .
Collapse
|
466
|
Seok JK, Lee SH, Kim MJ, Lee YM. MicroRNA-382 induced by HIF-1α is an angiogenic miR targeting the tumor suppressor phosphatase and tensin homolog. Nucleic Acids Res 2014; 42:8062-72. [PMID: 24914051 PMCID: PMC4081109 DOI: 10.1093/nar/gku515] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Recent studies have revealed that microRNAs (miRs) play important roles in the regulation of angiogenesis. In this study, we have characterized miR-382 upregulation by hypoxia and the functional relevance of miR-382 in tumor angiogenesis. miRs induced by hypoxia in MKN1 human gastric cancer cells were investigated using miRNA microarrays. We selected miR-382 and found that the expression of miR-382 was regulated by HIF-1α. Conditioned media (CM) from MKN1 cells transfected with a miR-382 inhibitor (antagomiR-382) under hypoxic conditions significantly decreased vascular endothelial cell (EC) proliferation, migration and tube formation. Algorithmic programs (Target Scan, miRanda and cbio) predicted that phosphatase and tensin homolog (PTEN) is a target gene of miR-382. Deletion of miR382-binding sequences in the PTEN mRNA 3′-untranslated region (UTR) diminished the luciferase reporter activity. Subsequent study showed that the overexpression of miR-382 or antagomiR-382 down- or upregulated PTEN and its downstream target AKT/mTOR signaling pathway, indicating that PTEN is a functional target gene of miR-382. In addition, PTEN inhibited miR-382-induced in vitro and in vivo angiogenesis as well as VEGF secretion, and the inhibition of miR-382 expression reduced xenograft tumor growth and microvessel density in tumors. Taken together, these results suggest that miR-382 induced by hypoxia promotes angiogenesis and acts as an angiogenic oncogene by repressing PTEN.
Collapse
Affiliation(s)
- Jin-Kyung Seok
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, 80 Daehak-ro, Buk-gu, Daegu, 702-701, Republic of Korea School of Life Sciences and Biotechnology, Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu, 702-701, Republic of Korea
| | - Sun Hee Lee
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, 80 Daehak-ro, Buk-gu, Daegu, 702-701, Republic of Korea
| | - Min Jung Kim
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, 80 Daehak-ro, Buk-gu, Daegu, 702-701, Republic of Korea School of Life Sciences and Biotechnology, Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu, 702-701, Republic of Korea
| | - You-Mie Lee
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, 80 Daehak-ro, Buk-gu, Daegu, 702-701, Republic of Korea School of Life Sciences and Biotechnology, Kyungpook National University, 80 Daehak-ro, Buk-gu, Daegu, 702-701, Republic of Korea
| |
Collapse
|
467
|
Gao Y, Jing M, Ge R, Zhou Z, Sun Y. Inhibition of hypoxia inducible factor 1α by siRNA-induced apoptosis in human retinoblastoma cells. J Biochem Mol Toxicol 2014; 28:394-9. [PMID: 24860939 DOI: 10.1002/jbt.21576] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Revised: 04/17/2014] [Accepted: 04/30/2014] [Indexed: 12/14/2022]
Abstract
Hypoxia, which activates the hypoxia inducible factor 1α (HIF-1α), is an essential feature of retinoblastoma (RB) and contributes to poor prognosis and resistance to conventional therapy. In this study, the effect of HIF-1α knockdown by small interfering RNA (siRNA) on cell proliferation, apoptosis, and apoptotic pathways of human Y-79 RB cells was first investigated. Exposure to hypoxia induced the increased expression of HIF-1α both in mRNA and protein levels. Then, knockdown of HIF-1α by siRNAHIF-1α resulted in inhibition of cell proliferation and induced cell apoptosis in human Y-79 RB cells under both normoxic and hypoxic conditions, with hypoxic conditions being more sensitive. Furthermore, knockdown of HIF-1α could enhance hypoxia-induced slight increase of Bax/Bcl-2 ratio and activate caspase-9 and caspase-3. These results together indicated that suppression of HIF-1α expression may be a promising strategy for the treatment of human RB in the future.
Collapse
Affiliation(s)
- Yu Gao
- Department of Ophthalmology, No. 411 Hospital of CPLA, Shanghai, 200081, People's Republic of China.
| | | | | | | | | |
Collapse
|
468
|
Tobler M, Henpita C, Bassett B, Kelley JL, Shaw JH. H2S exposure elicits differential expression of candidate genes in fish adapted to sulfidic and non-sulfidic environments. Comp Biochem Physiol A Mol Integr Physiol 2014; 175:7-14. [PMID: 24813672 DOI: 10.1016/j.cbpa.2014.04.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Revised: 03/26/2014] [Accepted: 04/17/2014] [Indexed: 11/16/2022]
Abstract
Disentangling the effects of plasticity, genetic variation, and their interactions on organismal responses to environmental stressors is a key objective in ecological physiology. We quantified the expression of five candidate genes in response to hydrogen sulfide (H2S) exposure in fish (Poecilia mexicana, Poeciliidae) from a naturally sulfide-rich environment as well as an ancestral, non-sulfidic population to test for constitutive and environmentally dependent population differences in gene expression patterns. Common garden raised individuals that had never encountered environmental H2S during their lifetime were subjected to short or long term H2S exposure treatments or respective non-sulfidic controls. The expression of genes involved in responses to H2S toxicity (cytochrome c oxidase, vascular endothelial growth factor, and cytochrome P450-2J6), H2S detoxification (sulfide:quinone oxidoreductase), and endogenous H2S production (cystathionine γ lyase) was determined in both gill and liver tissues by real time PCR. The results indicated complex changes in expression patterns that--depending on the gene--not only differed between organs and populations, but also on the type of H2S exposure. Populations differences, both constitutive and H2S exposure dependent (i.e., plastic), in gene expression were particularly evident for sulfide:quinone oxidoreductase, vascular endothelial growth factor, and to a lesser degree for cytochrome P450-2J6. Our study uncovered putatively adaptive modifications in gene regulation that parallel previously documented adaptive changes in phenotypic traits.
Collapse
Affiliation(s)
- Michael Tobler
- Department of Zoology, Oklahoma State University, 501 Life Sciences West, Stillwater, OK 74078, USA
| | - Chathurika Henpita
- Department of Zoology, Oklahoma State University, 501 Life Sciences West, Stillwater, OK 74078, USA
| | - Brandon Bassett
- Center for Veterinary Health Sciences, Oklahoma State University, 205 McElroy Hall, Stillwater, OK 74078, USA
| | - Joanna L Kelley
- School of Biological Sciences, Washington State University, PO Box 644236, Pullman, WA 99164, USA
| | - Jennifer H Shaw
- Department of Zoology, Oklahoma State University, 501 Life Sciences West, Stillwater, OK 74078, USA.
| |
Collapse
|
469
|
Hong WX, Hu MS, Esquivel M, Liang GY, Rennert RC, McArdle A, Paik KJ, Duscher D, Gurtner GC, Lorenz HP, Longaker MT. The Role of Hypoxia-Inducible Factor in Wound Healing. Adv Wound Care (New Rochelle) 2014; 3:390-399. [PMID: 24804159 DOI: 10.1089/wound.2013.0520] [Citation(s) in RCA: 260] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Accepted: 01/30/2014] [Indexed: 12/14/2022] Open
Abstract
Significance: Poor wound healing remains a significant health issue for a large number of patients in the United States. The physiologic response to local wound hypoxia plays a critical role in determining the success of the normal healing process. Hypoxia-inducible factor-1 (HIF-1), as the master regulator of oxygen homeostasis, is an important determinant of healing outcomes. HIF-1 contributes to all stages of wound healing through its role in cell migration, cell survival under hypoxic conditions, cell division, growth factor release, and matrix synthesis throughout the healing process. Recent Advances: Positive regulators of HIF-1, such as prolyl-4-hydroxylase inhibitors, have been shown to be beneficial in enhancing diabetic ischemic wound closure and are currently undergoing clinical trials for treatment of several human-ischemia-based conditions. Critical Issues: HIF-1 deficiency and subsequent failure to respond to hypoxic stimuli leads to chronic hypoxia, which has been shown to contribute to the formation of nonhealing ulcers. In contrast, overexpression of HIF-1 has been implicated in fibrotic disease through its role in increasing myofibroblast differentiation leading to excessive matrix production and deposition. Both positive and negative regulators of HIF-1 therefore provide important therapeutic targets that can be used to manipulate HIF-1 expression where an excess or deficiency in HIF-1 is known to correlate with pathogenesis. Future Directions: Targeting HIF-1 during wound healing has many important clinical implications for tissue repair. Counteracting the detrimental effects of excessive or deficient HIF-1 signaling by modulating HIF-1 expression may improve future management of poorly healing wounds.
Collapse
Affiliation(s)
- Wan Xing Hong
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California
- Department of Radiology, Stanford University School of Medicine, Stanford, California
- University of Central Florida College of Medicine, Orlando, Florida
| | - Michael S. Hu
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California
- Department of Surgery, John A. Burns School of Medicine, University of Hawai'i, Honolulu, Hawai'i
| | - Mikaela Esquivel
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California
| | - Grace Y. Liang
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Robert C. Rennert
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California
| | - Adrian McArdle
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California
| | - Kevin J. Paik
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California
| | - Dominik Duscher
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California
| | - Geoffrey C. Gurtner
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California
| | - H. Peter Lorenz
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California
| | - Michael T. Longaker
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
470
|
ZHANG ZHE, NIE FANGFEI, KANG CHUNFU, CHEN BIN, QIN ZELIAN, MA JIANXUN, MA YONGGUANG, ZHAO XIA. Increased periostin expression affects the proliferation, collagen synthesis, migration and invasion of keloid fibroblasts under hypoxic conditions. Int J Mol Med 2014; 34:253-61. [DOI: 10.3892/ijmm.2014.1760] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Accepted: 04/10/2014] [Indexed: 11/05/2022] Open
|
471
|
Campana L, Santarella F, Esposito A, Maugeri N, Rigamonti E, Monno A, Canu T, Del Maschio A, Bianchi ME, Manfredi AA, Rovere-Querini P. Leukocyte HMGB1 is required for vessel remodeling in regenerating muscles. THE JOURNAL OF IMMUNOLOGY 2014; 192:5257-64. [PMID: 24752445 DOI: 10.4049/jimmunol.1300938] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Signals of tissue necrosis, damage-associated molecular patterns (DAMPs), cause inflammation. Leukocytes migrating into injured tissues tonically release DAMPs, including the high mobility group box 1 protein (HMGB1). In the absence of suitable models, the relative role of DAMPs released because of necrosis or leukocyte activation has not, so far, been dissected. We have generated a mouse model lacking Hmgb1 in the hematopoietic system and studied the response to acute sterile injury of the skeletal muscle. Regenerating fibers are significantly less numerous at earlier time points and smaller at the end of the process. Leukocyte Hmgb1 licenses the skeletal muscle to react to hypoxia, to express angiopoietin-2, and to initiate angiogenesis in response to injury. Vascularization of the regenerating tissue is selectively jeopardized in the absence of leukocyte Hmgb1, revealing that it controls the nutrient and oxygen supply to the regenerating tissue. Altogether, our results reveal a novel nonredundant role for leukocyte Hmgb1 in the repair of injured skeletal muscle.
Collapse
Affiliation(s)
- Lara Campana
- Innate Immunity and Tissue Remodeling Unit, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Francesco Santarella
- Innate Immunity and Tissue Remodeling Unit, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Antonio Esposito
- Vita-Salute, San Raffaele University, 20132 Milan, Italy; Radiology Department, Centre for Experimental Imaging, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Norma Maugeri
- Autoimmunity and Vascular Inflammation Unit, San Raffaele Scientific Institute, 20132 Milan, Italy; and
| | - Elena Rigamonti
- Innate Immunity and Tissue Remodeling Unit, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Antonella Monno
- Innate Immunity and Tissue Remodeling Unit, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Tamara Canu
- Radiology Department, Centre for Experimental Imaging, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Alessandro Del Maschio
- Vita-Salute, San Raffaele University, 20132 Milan, Italy; Radiology Department, Centre for Experimental Imaging, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Marco E Bianchi
- Radiology Department, Centre for Experimental Imaging, San Raffaele Scientific Institute, 20132 Milan, Italy; Chromatin Dynamics Unit, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Angelo A Manfredi
- Vita-Salute, San Raffaele University, 20132 Milan, Italy; Autoimmunity and Vascular Inflammation Unit, San Raffaele Scientific Institute, 20132 Milan, Italy; and
| | - Patrizia Rovere-Querini
- Innate Immunity and Tissue Remodeling Unit, San Raffaele Scientific Institute, 20132 Milan, Italy; Vita-Salute, San Raffaele University, 20132 Milan, Italy;
| |
Collapse
|
472
|
Maquart F, Monboisse J. Extracellular matrix and wound healing. ACTA ACUST UNITED AC 2014; 62:91-5. [DOI: 10.1016/j.patbio.2014.02.007] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Accepted: 02/17/2014] [Indexed: 11/30/2022]
|
473
|
Kim BK, Kim BR, Lee HJ, Lee SA, Kim BJ, Kim H, Won YS, Shon WJ, Lee NR, Inn KS, Kim BJ. Tumor-suppressive effect of a telomerase-derived peptide by inhibiting hypoxia-induced HIF-1α-VEGF signaling axis. Biomaterials 2014; 35:2924-2933. [PMID: 24411674 DOI: 10.1016/j.biomaterials.2013.12.077] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Accepted: 12/20/2013] [Indexed: 01/13/2023]
Abstract
A reverse-transcriptase-subunit of telomerase (hTERT) derived peptide, GV1001, has been developed as a vaccine against various cancers. Previously, we have shown that GV1001 interacts with heat shock proteins (HSPs) and penetrates cell membranes to be localized in the cytoplasm. In this study, we have found that GV1001 lowered the level of intracellular and surface HSPs of various cancer cells. In hypoxic conditions, GV1001 treatment of cancer cells resulted in decreases of HSP90, HSP70, and HIF-1α. Subsequently, proliferation of cancer cells and synthesis of VEGF were significantly reduced by treatment using GV1001 in hypoxic conditions. In an experiment using a nude mouse xenograft model, GV1001 exerted a similar tumor suppressive effect, further confirming its anti-tumor efficacy. Higher apoptotic cell death, reduced proliferation of cells, and fewer blood vessels were observed in GV1001-treated tumors compared to control. In addition, significant reduction of Tie2+ CD11b+ monocytes, which were recruited by VEGF from tumor cells and play a critical role in angiogenesis, was observed in GV1001-treated tumors. Collectively, the results suggest that GV1001 possesses potential therapeutic efficacy in addition to its ability to induce anti-cancer immune responses by suppressing both HSP70 and HSP90.
Collapse
Affiliation(s)
- Bu-Kyung Kim
- Department of Microbiology and Immunology, Liver Research Institute, Cancer Research Institute and SNUMRC, College of Medicine, Seoul National University, 28 Yongon-dong, Chongno-gu, Seoul 110-799, Republic of Korea
| | - Bo-Ram Kim
- Department of Microbiology and Immunology, Liver Research Institute, Cancer Research Institute and SNUMRC, College of Medicine, Seoul National University, 28 Yongon-dong, Chongno-gu, Seoul 110-799, Republic of Korea
| | - Hyun-Joo Lee
- Department of Microbiology and Immunology, Liver Research Institute, Cancer Research Institute and SNUMRC, College of Medicine, Seoul National University, 28 Yongon-dong, Chongno-gu, Seoul 110-799, Republic of Korea
| | - Seoung-Ae Lee
- Department of Microbiology and Immunology, Liver Research Institute, Cancer Research Institute and SNUMRC, College of Medicine, Seoul National University, 28 Yongon-dong, Chongno-gu, Seoul 110-799, Republic of Korea
| | - Byoung-Jun Kim
- Department of Microbiology and Immunology, Liver Research Institute, Cancer Research Institute and SNUMRC, College of Medicine, Seoul National University, 28 Yongon-dong, Chongno-gu, Seoul 110-799, Republic of Korea
| | - Hong Kim
- Department of Microbiology and Immunology, Liver Research Institute, Cancer Research Institute and SNUMRC, College of Medicine, Seoul National University, 28 Yongon-dong, Chongno-gu, Seoul 110-799, Republic of Korea
| | - Yu-Sub Won
- Department of Microbiology and Immunology, Liver Research Institute, Cancer Research Institute and SNUMRC, College of Medicine, Seoul National University, 28 Yongon-dong, Chongno-gu, Seoul 110-799, Republic of Korea
| | - Won-Jun Shon
- Department of Conservative Dentistry, Dental Research Institute and School of Dentistry, Seoul National University, Seoul, Republic of Korea
| | - Na-Rae Lee
- Department of Pharmaceutical Science, College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 130-701, Republic of Korea
| | - Kyung-Soo Inn
- Department of Pharmaceutical Science, College of Pharmacy, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 130-701, Republic of Korea.
| | - Bum-Joon Kim
- Department of Microbiology and Immunology, Liver Research Institute, Cancer Research Institute and SNUMRC, College of Medicine, Seoul National University, 28 Yongon-dong, Chongno-gu, Seoul 110-799, Republic of Korea.
| |
Collapse
|
474
|
Zheng J, Nagda DA, Lajud SA, Kumar S, Mouchli A, Bezpalko O, O'Malley BW, Li D. Biliverdin's regulation of reactive oxygen species signalling leads to potent inhibition of proliferative and angiogenic pathways in head and neck cancer. Br J Cancer 2014; 110:2116-22. [PMID: 24569474 PMCID: PMC3992514 DOI: 10.1038/bjc.2014.98] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Revised: 01/29/2014] [Accepted: 01/30/2014] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND In this study, we evaluate whether the use of biliverdin (BV), a natural non-toxic antioxidant product of haeme catabolism, can suppress head and neck squamous cell carcinoma (HNSCC) cell proliferation and improve the tumour survival both in vitro and in vivo. Furthermore, we investigate whether this therapeutic outcome relies on BV's potent antioxidant effect on reactive oxygen species (ROS)-mediated signalling. METHODS Two well-characterised HNSCC cell lines and a mouse model with human HNSCC were used for this study. In vitro, the effect of BV on ROS was assayed. Subsequently, critical regulatory proteins involved in growth, antiapoptotic, and angiogenic pathways were investigated by western blot analysis. In addition, the antiproliferative effect of BV was also evaluated using the clonogenic assay. Moreover, tumour growth inhibition was assessed using a mouse model with HNSCC. RESULTS Biliverdin treatment resulted in decreased ROS, leading to suppression of proliferation and angiogenesis pathways of HNSCC, significantly decreasing the expression and phosphorylation of oncogenic factors such as epidermal growth factor receptor (EGFR), phosphorylation of Akt, and expression of angiogenic marker and transcription factor, hypoxia-inducible factor1-α (HIF1-α). Furthermore, this downregulation of ROS by BV led to a significant suppression of tumour growth in vivo. CONCLUSIONS Our study demonstrates the efficacy of a novel therapeutic approach using BV as an antitumour agent against HNSCC through its effect on EGFR/Akt and HIF1-α/angiogenesis signal transduction pathways. Our findings indicate that BV's inhibitory effect on these tumorigenic pathways relies on its antioxidant effect, and may extend its therapeutic potential to other solid cancers.
Collapse
Affiliation(s)
- J Zheng
- 1] Department of Otorhinolaryngology-Head and Neck Surgery, University of Pennsylvania Perelman School of Medicine, 415 Curie Boulevard, Clinical Research Building, Philadelphia, PA, USA [2] Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - D A Nagda
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Pennsylvania Perelman School of Medicine, 415 Curie Boulevard, Clinical Research Building, Philadelphia, PA, USA
| | - S A Lajud
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Pennsylvania Perelman School of Medicine, 415 Curie Boulevard, Clinical Research Building, Philadelphia, PA, USA
| | - S Kumar
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Pennsylvania Perelman School of Medicine, 415 Curie Boulevard, Clinical Research Building, Philadelphia, PA, USA
| | - A Mouchli
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Pennsylvania Perelman School of Medicine, 415 Curie Boulevard, Clinical Research Building, Philadelphia, PA, USA
| | - O Bezpalko
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Pennsylvania Perelman School of Medicine, 415 Curie Boulevard, Clinical Research Building, Philadelphia, PA, USA
| | - B W O'Malley
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Pennsylvania Perelman School of Medicine, 415 Curie Boulevard, Clinical Research Building, Philadelphia, PA, USA
| | - D Li
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Pennsylvania Perelman School of Medicine, 415 Curie Boulevard, Clinical Research Building, Philadelphia, PA, USA
| |
Collapse
|
475
|
In Vivo Evaluation of Wound Bed Reaction and Graft Performance After Cold Skin Graft Storage. J Burn Care Res 2014; 35:e187-96. [DOI: 10.1097/bcr.0b013e3182a226df] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
|
476
|
Yang SL, Liu LP, Jiang JX, Xiong ZF, He QJ, Wu C. The correlation of expression levels of HIF-1α and HIF-2α in hepatocellular carcinoma with capsular invasion, portal vein tumor thrombi and patients' clinical outcome. Jpn J Clin Oncol 2013; 44:159-67. [PMID: 24374892 DOI: 10.1093/jjco/hyt194] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
OBJECTIVES The roles of hypoxia-inducible factor-1α and hypoxia-inducible factor-2α in the development of hepatocellular carcinoma have not been fully elucidated. Here, we aim to uncover the relationship between the prognosis of hepatocellular carcinoma patients and the expression of hypoxia-inducible factor-1α and hypoxia-inducible factor-2α in tumor tissues. METHODS The protein levels of hypoxia-inducible factor-1α and hypoxia-inducible factor-2α were detected by immunohistochemistry on paraffin sections of 126 paired hepatocellular carcinoma tissue and peritumoral tissue samples. The mRNA levels of them were detected by quantitative real-time polymerase chain reaction. RESULTS High expression of hypoxia-inducible factor-1α was found in 57.1% (72/126) of tumor specimens, compared with 5.6% (7/126) in peritumoral tissues, while high expression of hypoxia-inducible factor-2α was found in only 13.5% (17/126) of tumors, compared with 47.6% (60/126) of peritumoral tissues. There was high expression of hypoxia-inducible factor-1α protein in hepatocellular carcinoma tissues closely associated with capsular infiltration and portal vein invasion, and thus lower overall survival and disease-free survival of hepatocellular carcinoma patients (P < 0.05). No significant association has been found between the expression of hypoxia-inducible factor-2α protein and capsular infiltration, portal vein invasion, overall survival and disease-free survival (P > 0.05). However, patients with high expression of both hypoxia-inducible factor-1α and hypoxia-inducible factor-2α have a significantly worse outcome than patients with low expression of both hypoxia-inducible factor-1α and hypoxia-inducible factor-2α (P < 0.05). CONCLUSIONS The discordant results on expression of hypoxia-inducible factor-1α and hypoxia-inducible factor-2α suggest that these two proteins are differentially regulated in vivo, thus reflecting distinctive protein expression and stabilization mechanisms. The association between hypoxia-inducible factor-1α expression and unfavorable outcome indicates the importance of using hypoxia-inducible factor-1α as a treatment target in hepatocellular carcinoma.
Collapse
Affiliation(s)
- Sheng-Li Yang
- *Department of Hepatobiliary Surgery, Affiliated Hospital of Guiyang Medical College, Guiyang, Guizhou 550001, China.
| | | | | | | | | | | |
Collapse
|
477
|
Shi D, Xie F, Zhang Y, Tian Y, Chen W, Fu L, Wang J, Guo W, Kang T, Huang W, Deng W. TFAP2A Regulates Nasopharyngeal Carcinoma Growth and Survival by Targeting HIF-1α Signaling Pathway. Cancer Prev Res (Phila) 2013; 7:266-77. [DOI: 10.1158/1940-6207.capr-13-0271] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
478
|
Chen Z, Wu C, Gu W, Klein T, Crawford R, Xiao Y. Osteogenic differentiation of bone marrow MSCs by β-tricalcium phosphate stimulating macrophages via BMP2 signalling pathway. Biomaterials 2013; 35:1507-18. [PMID: 24268199 DOI: 10.1016/j.biomaterials.2013.11.014] [Citation(s) in RCA: 238] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Accepted: 11/03/2013] [Indexed: 02/02/2023]
Abstract
Immune reactions play important roles in determining the in vivo fate of bone substitute materials, either in new bone formation or inflammatory fibrous tissue encapsulation. The paradigm for the development of bone substitute materials has been shifted from inert to immunomodulatory materials, emphasizing the importance of immune cells in the material evaluation. Macrophages, the major effector cells in the immune reaction to implants, are indispensable for osteogenesis and their heterogeneity and plasticity render macrophages a primer target for immune system modulation. However, there are very few reports about the effects of macrophages on biomaterial-regulated osteogenesis. In this study, we used β-tricalcium phosphate (β-TCP) as a model biomaterial to investigate the role of macrophages on the material stimulated osteogenesis. The macrophage phenotype switched to M2 extreme in response to β-TCP extracts, which was related to the activation of calcium-sensing receptor (CaSR) pathway. Bone morphogenetic protein 2 (BMP2) was also significantly upregulated by the β-TCP stimulation, indicating that macrophage may participate in the β-TCP stimulated osteogenesis. Interestingly, when macrophage-conditioned β-TCP extracts were applied to bone marrow mesenchymal stem cells (BMSCs), the osteogenic differentiation of BMSCs was significantly enhanced, indicating the important role of macrophages in biomaterial-induced osteogenesis. These findings provided valuable insights into the mechanism of material-stimulated osteogenesis, and a strategy to optimize the evaluation system for the in vitro osteogenesis capacity of bone substitute materials.
Collapse
Affiliation(s)
- Zetao Chen
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, 60 Musk Ave, Kelvin Grove, Brisbane, Queensland 4059, Australia
| | - Chengtie Wu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, 1295 Dingxi Road, Shanghai 200050, People's Republic of China.
| | - Wenyi Gu
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, 60 Musk Ave, Kelvin Grove, Brisbane, Queensland 4059, Australia; Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Corner College and Cooper Rds, Brisbane, Queensland 4072, Australia
| | - Travis Klein
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, 60 Musk Ave, Kelvin Grove, Brisbane, Queensland 4059, Australia
| | - Ross Crawford
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, 60 Musk Ave, Kelvin Grove, Brisbane, Queensland 4059, Australia
| | - Yin Xiao
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, 60 Musk Ave, Kelvin Grove, Brisbane, Queensland 4059, Australia.
| |
Collapse
|
479
|
Li YN, Xi MM, Guo Y, Hai CX, Yang WL, Qin XJ. NADPH oxidase-mitochondria axis-derived ROS mediate arsenite-induced HIF-1α stabilization by inhibiting prolyl hydroxylases activity. Toxicol Lett 2013; 224:165-74. [PMID: 24188932 DOI: 10.1016/j.toxlet.2013.10.029] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2013] [Revised: 10/24/2013] [Accepted: 10/25/2013] [Indexed: 12/17/2022]
Abstract
Arsenic exposure has been shown to induce hypoxia inducible factor 1α (HIF-1α) accumulation, however the underlying mechanism remains unknown. In the present study, we tested the hypothesis that arsenic exposure triggered the interaction between NADPH oxidase and mitochondria to promote reactive oxygen species (ROS) production, which inactivate prolyl hydroxylases (PHDs) activity, leading to the stabilization of HIF-1α protein. Exposure of human immortalized liver cell line HL-7702 cells to arsenite induced HIF-1α accumulation in a dose-dependent manner, which was abolished by SOD mimetic MnTMPyP. Inhibition of NADPH oxidase with diphenyleneiodonium chloride (DPI) or inhibition of mitochondrial respiratory chain with rotenone significantly blocked arsenite-induced ROS production, and the mitochondria appeared to be the major source of ROS production. Arsenite treatment inhibited HIF-1α hydroxylation by prolyl hydroxylases (PHDs) and increased HIF-1α stabilization, but did not affect HIF-1α mRNA expression and Akt activation. Supplementation of ascorbate or Fe(II) completely abolished arsenite-induced PHDs inhibition and HIF-1α stabilization. In conclusion, these results define a unique mechanism of HIF-1α accumulation following arsenic exposure, that is, arsenic activates NADPH oxidase-mitochondria axis to produce ROS, which deplete intracellular ascorbate and Fe(II) to inactivate PHDs, leading to HIF-1α stabilization.
Collapse
Affiliation(s)
- Ying-Na Li
- Department of Geriatrics, The Second Affiliated Hospital of Medicine School, The Xi'an Jiaotong University, Xi'an 710004, China
| | | | | | | | | | | |
Collapse
|
480
|
Zheng M, Jiang J, Tang YL, Liang XH. Oncogene and non-oncogene addiction in inflammation-associated cancers. Future Oncol 2013; 9:561-73. [PMID: 23560378 DOI: 10.2217/fon.12.202] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Many cancers originate in tissues that are chronically inflamed, and the inflammatory microenvironment is considered to promote the progression of malignancy, including initiation, growth, angiogenesis, invasion and metastasis. The molecular mechanism of inflammation-induced progression of cancers has been widely discussed. Oncogene and non-oncogene addiction have been proposed as two distinct but complementary theories to explain the initiation and development of cancers. Furthermore, they also play a role in cancer-associated inflammation. A solid understanding of oncogene and non-oncogene addiction in cancer-associated inflammatory microenvironments will help to exploit cancer drug targets for cancer prevention and clinical treatment.
Collapse
Affiliation(s)
- Min Zheng
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, No. 14, Sec. 3, Renminnan Road, Chengdu Sichuan 610041, People's Republic of China
| | | | | | | |
Collapse
|
481
|
HIF1α is required for osteoclast activation by estrogen deficiency in postmenopausal osteoporosis. Proc Natl Acad Sci U S A 2013; 110:16568-73. [PMID: 24023068 DOI: 10.1073/pnas.1308755110] [Citation(s) in RCA: 152] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
In women, estrogen deficiency after menopause frequently accelerates osteoclastic bone resorption, leading to osteoporosis, the most common skeletal disorder. However, mechanisms underlying osteoporosis resulting from estrogen deficiency remain largely unknown. Here we show that in bone-resorbing osteoclasts, estrogen-dependent destabilization of hypoxia-inducible factor 1 alpha (HIF1α), which is unstable in the presence of oxygen, plays a pivotal role in promoting bone loss in estrogen-deficient conditions. In vitro, HIF1α was destabilized by estrogen treatment even in hypoxic conditions, and estrogen loss in ovariectomized (Ovx) mice stabilized HIF1α in osteoclasts and promoted their activation and subsequent bone loss in vivo. Osteoclast-specific HIF1α inactivation antagonized bone loss in Ovx mice and osteoclast-specific estrogen receptor alpha deficient mice, both models of estrogen-deficient osteoporosis. Oral administration of a HIF1α inhibitor protected Ovx mice from osteoclast activation and bone loss. Thus, HIF1α represents a promising therapeutic target in osteoporosis.
Collapse
|
482
|
Ling S, Birnbaum Y, Nanhwan MK, Thomas B, Bajaj M, Ye Y. MicroRNA-dependent cross-talk between VEGF and HIF1α in the diabetic retina. Cell Signal 2013; 25:2840-7. [PMID: 24018047 DOI: 10.1016/j.cellsig.2013.08.039] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Accepted: 08/30/2013] [Indexed: 12/13/2022]
Abstract
Both HIF1α (hypoxia-inducible factor alpha) and VEGF (vascular endothelial growth factor) are implicated in the pathogenesis of diabetic retinopathy (DR). Competitive endogenous RNAs (ceRNAs) are messenger RNA (mRNA) molecules that affect each other expression through competition for their shared microRNAs (miRNA). However, little is known about the role of ceRNAs in DR. We assess whether the expression of HIF1α and VEGF in DR is interdependent through sequestration of common miRNAs. We used bioinformatics to identify potential miRNAs that affect both genes and validated the interdependence of the genes by silencing or overexpression of the genes and assessed the luciferase-HIF1α 3'UTR activity. We found that HIF1α and VEGF are targeted by 12 common miRNAs. Silencing either HIF1α or VEGF increased the availabilities of the shared miRNAs, therefore suppressed the luciferase-HIF1α 3'UTR activity, whereas over-expressing HIF1α or VEGF increased the luciferase activity. HIF1α was co-expressed with VEGF in-vivo and in-vitro in DR models. Silencing HIF1α transcripts resulted in a significant reduction in VEGF protein levels and vice versa. This interdependence was miRNA- and 3'UTR-dependent, as silencing Dicer abolished the interdependence. Over-expression of a common miRNA (miR-106a) significantly reduced the expression of HIF1α and VEGF and prevented high glucose-induced increased permeability. There is a cross-talk between HIF1α and VEGF through interactions with their common miRNAs. miRNA based therapy can affect the expression of both HIF1α and VEGF and may represent a therapeutic potential for the treatment of DR.
Collapse
Affiliation(s)
- Shukuan Ling
- The Department of Biochemistry and Molecular Biology, University of Texas Medical Branch; Galveston, TX, USA; School of Life Science and Technology, Harbin Institute of Technology, Harbin, China; State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | | | | | | | | | | |
Collapse
|
483
|
Meidan R, Klipper E, Zalman Y, Yalu R. The role of hypoxia-induced genes in ovarian angiogenesis. Reprod Fertil Dev 2013; 25:343-50. [PMID: 22950963 DOI: 10.1071/rd12139] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2012] [Accepted: 07/26/2012] [Indexed: 01/23/2023] Open
Abstract
The hypoxic microenvironment that occurs in fast-growing tissue such as the corpus luteum (CL) is a major contributor to its ability to survive via the induction of an intricate vascular network. Cellular responses to hypoxia are mediated by hypoxia-inducible factor-1 (HIF-1), an oxygen-regulated transcriptional activator. HIF-1, a heterodimer consisting of a constitutively-expressed β subunit and an oxygen-regulated α subunit, binds to the hypoxia responsive element (HRE) present in the promoter regions of responsive genes. This review summarises evidence for the involvement of hypoxia and HIF-1α in CL development and function. Special emphasis is given to hypoxia-induced, luteal cell-specific expression of multiple genes (vascular endothelial growth factor A (VEGFA), fibroblast growth factor 2 (FGF-2), prokineticin receptor 2 (PK-R2), stanniocalcin 1 (STC-1) and endothelin 2 (EDN-2) that participate in the angiogenic process during CL formation.
Collapse
Affiliation(s)
- Rina Meidan
- Department of Animal Sciences, The Hebrew University of Jerusalem, The Robert H. Smith Faculty of Agriculture, Food and Environment, Rehovot 76100, Israel.
| | | | | | | |
Collapse
|
484
|
Shin JM, Jeong YJ, Cho HJ, Park KK, Chung IK, Lee IK, Kwak JY, Chang HW, Kim CH, Moon SK, Kim WJ, Choi YH, Chang YC. Melittin suppresses HIF-1α/VEGF expression through inhibition of ERK and mTOR/p70S6K pathway in human cervical carcinoma cells. PLoS One 2013; 8:e69380. [PMID: 23936001 PMCID: PMC3720276 DOI: 10.1371/journal.pone.0069380] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Accepted: 06/08/2013] [Indexed: 01/01/2023] Open
Abstract
OBJECTIVE Melittin (MEL), a major component of bee venom, has been associated with various diseases including arthritis, rheumatism and various cancers. In this study, the anti-angiogenic effects of MEL in CaSki cells that were responsive to the epidermal growth factor (EGF) were examined. METHODOLOGY/PRINCIPAL FINDINGS MEL decreased the EGF-induced hypoxia-inducible factor-1α (HIF-1α) protein and significantly regulated angiogenesis and tumor progression. We found that inhibition of the HIF-1α protein level is due to the shortened half-life by MEL. Mechanistically, MEL specifically inhibited the EGF-induced HIF-1α expression by suppressing the phosphorylation of ERK, mTOR and p70S6K. It also blocked the EGF-induced DNA binding activity of HIF-1α and the secretion of the vascular endothelial growth factor (VEGF). Furthermore, the chromatin immunoprecipitation (ChIP) assay revealed that MEL reduced the binding of HIF-1α to the VEGF promoter HRE region. The anti-angiogenesis effects of MEL were confirmed through a matrigel plus assay. CONCLUSIONS MEL specifically suppressed EGF-induced VEGF secretion and new blood vessel formation by inhibiting HIF-1α. These results suggest that MEL may inhibit human cervical cancer progression and angiogenesis by inhibiting HIF-1α and VEGF expression.
Collapse
Affiliation(s)
- Jae-Moon Shin
- Research Institute of Biomedical Engineering and Department of Medicine, Catholic University of Daegu School of Medicine, Daegu, Republic of Korea
| | - Yun-Jeong Jeong
- Research Institute of Biomedical Engineering and Department of Medicine, Catholic University of Daegu School of Medicine, Daegu, Republic of Korea
| | - Hyun-Ji Cho
- Research Institute of Biomedical Engineering and Department of Medicine, Catholic University of Daegu School of Medicine, Daegu, Republic of Korea
| | - Kwan-Kyu Park
- Research Institute of Biomedical Engineering and Department of Medicine, Catholic University of Daegu School of Medicine, Daegu, Republic of Korea
| | - Il-Kyung Chung
- Department of Biotechnology, Catholic University of Daegu, Gyeongsan, Republic of Korea
| | - In-Kyu Lee
- Department of Internal Medicine, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Jong-Young Kwak
- Department of Biochemistry, Dong-A University College of Medicine, Busan, Republic of Korea
| | - Hyeun-Wook Chang
- College of Pharmacy, Yeungnam University, Gyeongsan, Republic of Korea
| | - Cheorl-Ho Kim
- Department of Biological Science, Sungkyunkwan University, Suwon, Republic of Korea
| | - Sung-Kwon Moon
- Department of Food Science and Technology, Chung-Ang University, Ansung, Republic of Korea
| | - Wun-Jae Kim
- Personalized Tumor Engineering Research Center, Department of Urology, Chungbuk National University, Cheongju, Republic of Korea
| | - Yung-Hyun Choi
- Department of Biochemistry, College of Oriental Medicine, Dongeui University, Busan, Republic of Korea
| | - Young-Chae Chang
- Research Institute of Biomedical Engineering and Department of Medicine, Catholic University of Daegu School of Medicine, Daegu, Republic of Korea
| |
Collapse
|
485
|
Allon I, Kaplan I, Allon DM, Vered H, Shlomi B, Hirshberg A. HIF-1α, VEGF, and EGFR: contributing factors in the pathogenesis of necrotizing sialometaplasia. Oral Dis 2013; 20:440-5. [PMID: 23837804 DOI: 10.1111/odi.12149] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Revised: 05/22/2013] [Accepted: 06/16/2013] [Indexed: 11/27/2022]
Abstract
OBJECTIVES Necrotizing sialometaplasia (NS) is an uncommon reactive lesion involving the minor salivary glands. This study aimed to investigate the expression of hypoxia-inducible factor alpha (HIF-1α), vascular endothelial growth factor (VEGF), and epithelial growth factor receptor (EGFR) in the pathogenesis of NS. METHODS Paraffin-embedded tissue sections from 10 cases of NS were immunohistochemically stained for HIF-1α, VEGF, and EGFR. A semiquantitative morphometric analysis was performed and compared with normal palatal salivary glands and traumatic ulcerations. RESULTS Hypoxia-inducible factor alpha staining was observed in most elements of the affected area, the acini and ducts of the involved salivary glands as well as in the inflammatory infiltrate, the endothelial cells, and stromal cells. HIF-1α was almost absent in the control glands (P < 0.0001). VEGF staining was positive in the stromal capillaries and in the inflammatory infiltrate. The expression was higher in cases of NS compared with the normal salivary glands (P < 0.001). EGFR was expressed in the surface epithelium, the pseudo-epitheliomatous hyperplasia, and the islands of squamous metaplasia. VEGF expression in traumatic ulcerations was lower than that in cases of NS. CONCLUSION This study provides molecular evidence to the role of hypoxia in NS; HIF-1α, the main regulator of hypoxia, was expressed in the infarcted salivary glands, EGFR in the metaplastic epithelium and VEGF in the stromal capillaries, all three components are the key factors induced by hypoxia.
Collapse
Affiliation(s)
- I Allon
- Department of Oral Pathology & Oral Medicine, Goldschleger School of Dental Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | | | | | | | | |
Collapse
|
486
|
Nagaraju GP, Zafar SF, El-Rayes BF. Pleiotropic effects of genistein in metabolic, inflammatory, and malignant diseases. Nutr Rev 2013; 71:562-72. [PMID: 23865800 DOI: 10.1111/nure.12044] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Genistein is a soy-derived biologically active isoflavone that exhibits diverse health-promoting effects. An increasing body of evidence shows that genistein influences lipid homeostasis and insulin resistance, counteracts inflammatory cytokines, and possesses antidiabetic properties. Genistein also impedes cancer progression by promoting apoptosis, inducing cell cycle arrest, modulating intracellular signaling pathways, and inhibiting angiogenesis and metastasis of neoplastic cells. This review summarizes the pleiotropic functions of genistein in common health disorders such as metabolic syndrome, chronic inflammatory diseases, and cancer. In the current era of uncontrolled health expenditure, a focus on the clinical development of nutritional agents with the capacity to prevent a variety of common health disorders is needed. As a micronutrient that exerts multifaceted effects ranging from antidiabetic to anticarcinogenic functions, genistein should be clinically developed further for use in the prevention and treatment of a variety of health disorders.
Collapse
Affiliation(s)
- Ganji Purnachandra Nagaraju
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
| | | | | |
Collapse
|
487
|
Jia D, Tang B, Shi Y, Wang J, Sun Z, Chen Z, Zhang L, Xia K, Jiang H. A deletion mutation of the VHL gene associated with a patient with sporadic von Hippel-Lindau disease. J Clin Neurosci 2013; 20:842-7. [DOI: 10.1016/j.jocn.2012.06.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2012] [Revised: 05/26/2012] [Accepted: 06/10/2012] [Indexed: 11/25/2022]
|
488
|
Stathmin Regulates Hypoxia-Inducible Factor-1α Expression through the Mammalian Target of Rapamycin Pathway in Ovarian Clear Cell Adenocarcinoma. ISRN PHARMACOLOGY 2013; 2013:279593. [PMID: 23819061 PMCID: PMC3683482 DOI: 10.1155/2013/279593] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Accepted: 05/13/2013] [Indexed: 01/17/2023]
Abstract
Stathmin, a microtubule-destabilizing phosphoprotein, is highly expressed in ovarian cancer, but the pathophysiological significance of this protein in ovarian carcinoma cells remains poorly understood. This study reports the involvement of stathmin in the mTOR/HIF-1α/VEGF pathway in ovarian clear cell adenocarcinoma (CCA) during hypoxia. HIF-1α protein and VEGF mRNA levels were markedly elevated in RMG-1 cells, a CCA cell line, cultured under hypoxic conditions. Rapamycin, an inhibitor of mTOR complex 1, reduced the level of HIF-1α and blocked phosphorylation of ribosomal protein S6 kinase 1 (S6K), a transcriptional regulator of mTOR, demonstrating that hypoxia activates mTOR/S6K/HIF-1α signaling in CCA. Furthermore, stathmin knockdown inhibited hypoxia-induced HIF-1α and VEGF expression and S6K phosphorylation. The silencing of stathmin expression also reduced Akt phosphorylation, a critical event in the mTOR/HIF-1α/VEGF signaling pathway. By contrast, stathmin overexpression upregulated hypoxia-induced HIF-1α and VEGF expression in OVCAR-3 cells, another CCA cell line. In addition, suppression of Akt activation by wortmannin, a phosphoinositide 3-kinase (PI3K) inhibitor, decreased HIF-1α and VEGF expression. These results illustrate that regulation of HIF-1α through the PI3K/Akt/mTOR pathway is controlled by stathmin in CCA. Our findings point to a new mechanism of stathmin regulation during ovarian cancer.
Collapse
|
489
|
PF-04691502 triggers cell cycle arrest, apoptosis and inhibits the angiogenesis in hepatocellular carcinoma cells. Toxicol Lett 2013; 220:150-6. [PMID: 23639247 DOI: 10.1016/j.toxlet.2013.04.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Revised: 04/12/2013] [Accepted: 04/22/2013] [Indexed: 12/21/2022]
Abstract
Hepatocellular carcinoma (HCC) is a major cause of morbidity and mortality in the world. The aim of the present study is to determine the antitumor effect of PF-04691502, a potent inhibitor of PI3K and mTOR kinases, on the apoptosis and angiogenesis of the hepatoma cancer cells. Our results indicate that treatment of cancer cells with PF-04691502 reduces cell viability and inhibits cell growth in a dose-dependent manner. PF-04691502 triggers apoptosis via a mitochondrial pathway, accompanied by activation of caspase-3, caspase-9, and poly(ADP-ribose) polymerase (PARP). Pre-treatment of hepatoma cells with the caspase-3 inhibitor (z-DEVD-fmk) blocks the PF-04691502-induced death of these cells. In addition, growth factors-induced tube formation and the migration of HUVECs are markedly inhibited by PF-04691502 treatment. The mechanisms of anti-angiogenesis of PF-04691502 are associated with inhibiting the expression of VEGF and HIF-1α. Based on the overall results, we suggest that PF-04691502 reduces hepatocellular carcinoma cell viability, induces cell apoptosis, and inhibits cell growth and tumor angiogenesis, implicating its potential therapeutic value in the treatment of HCC.
Collapse
|
490
|
Tafani M, Pucci B, Russo A, Schito L, Pellegrini L, Perrone GA, Villanova L, Salvatori L, Ravenna L, Petrangeli E, Russo MA. Modulators of HIF1α and NFkB in Cancer Treatment: Is it a Rational Approach for Controlling Malignant Progression? Front Pharmacol 2013; 4:13. [PMID: 23408731 PMCID: PMC3569619 DOI: 10.3389/fphar.2013.00013] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2012] [Accepted: 01/23/2013] [Indexed: 01/17/2023] Open
Abstract
HIF1α and NFkB are two transcription factors very frequently activated in tumors and involved in tumor growth, progression, and resistance to chemotherapy. In fact, HIF1α and NFkB together regulate transcription of over a thousand genes that, in turn, control vital cellular processes such as adaptation to the hypoxia, metabolic reprograming, inflammatory reparative response, extracellular matrix digestion, migration and invasion, adhesion, etc. Because of this wide involvement they could control in an integrated manner the origin of the malignant phenotype. Interestingly, hypoxia and inflammation have been sequentially bridged in tumors by the discovery that alarmin receptors genes such as RAGE, P2X7, and some TLRs, are activated by HIF1α; and that, in turn, alarmin receptors strongly activate NFkB and proinflammatory gene expression, evidencing all the hallmarks of the malignant phenotype. Recently, a large number of drugs have been identified that inhibit one or both transcription factors with promising results in terms of controlling tumor progression. In addition, many of these molecules are natural compounds or off-label drugs already used to cure other pathologies. Some of them are undergoing clinical trials and soon they will be used alone or in combination with standard anti-tumoral agents to achieve a better treatment of tumors with reduction of metastasis formation and, more importantly, with a net increase in survival. This review highlights the central role of HIF1α activated in hypoxic regions of the tumor, of NFkB activation and proinflammatory gene expression in transformed cells to understand their progression toward malignancy. Different molecules and strategies to inhibit these transcription factors will be reviewed. Finally, the central role of a new class of deacetylases called Sirtuins in regulating HIF1α and NFkB activity will be outlined.
Collapse
Affiliation(s)
- Marco Tafani
- Department of Experimental Medicine, Sapienza University of Rome Rome, Italy ; Laboratory of Molecular and Cellular Pathology - Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele Pisana Rome, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
491
|
Tanaka M, Masaki Y, Tanaka K, Miyazaki M, Kato M, Sugimoto R, Nakamura K, Aishima S, Shirabe K, Nakamuta M, Enjoji M, Kotoh K, Takayanagi R. Reduction of fatty acid oxidation and responses to hypoxia correlate with the progression of de-differentiation in HCC. Mol Med Rep 2013; 7:365-70. [PMID: 23178736 DOI: 10.3892/mmr.2012.1201] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Accepted: 11/16/2012] [Indexed: 12/16/2022] Open
Abstract
The prognosis of patients with hepatocellular carcinoma (HCC) may be improved by novel treatments focusing on the characteristic metabolic changes of this disease. Therefore, we analyzed the biological interactions of metabolic features with the degree of tumor differentiation and the level of malignant potential in 41 patients with completely resectable HCC. The expression levels in resected samples of mRNAs encoded by genes related to tumor metabolism and metastasis were investigated, and the correlation between these expression levels and degrees of differentiation was analyzed. Of the 41 patients, 2 patients had grade I, 27 had grade II, and 12 had grade III tumors. Reductions in the levels of 3-hydroxyacyl-CoA dehydrogenase (HADHA) and acyl-CoA oxidase (ACOX)-2 mRNAs, and increases in pyruvate kinase isoenzyme type M2 (PKM2) mRNA were significantly correlated with the progression of de-differentiation. Analysis of partial correlation coefficients showed that the level of PKM2 mRNA expression was significantly correlated with those of pro-angiogenic genes, vascular endothelial growth factor (VEGF) and ETS-1. Moreover, the levels of VEGF-A and ETS-1 mRNA expression were independently correlated with that of the epithelial-mesenchymal transition (EMT)‑related gene SNAIL. These findings suggest that reductions in fatty acid oxidation and responses to hypoxia may affect the progression of malignant phenotypes in HCC.
Collapse
Affiliation(s)
- Masatake Tanaka
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Higashi-ku, Fukuoka 812-8582, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
492
|
Roles for PI3K/AKT/PTEN Pathway in Cell Signaling of Nonalcoholic Fatty Liver Disease. ISRN ENDOCRINOLOGY 2013; 2013:472432. [PMID: 23431468 PMCID: PMC3570922 DOI: 10.1155/2013/472432] [Citation(s) in RCA: 128] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2012] [Accepted: 01/02/2013] [Indexed: 12/15/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common form of liver pathologies and is associated with obesity and the metabolic syndrome, which represents a range of fatty liver diseases associated with an increased risk of type 2 diabetes. Molecular mechanisms underlying how to make transition from simple fatty liver to nonalcoholic steatohepatitis (NASH) are not well understood. However, accumulating evidence indicates that deregulation of the phosphatidylinositol 3-kinase (PI3K)/AKT pathway in hepatocytes is a common molecular event associated with metabolic dysfunctions including obesity, metabolic syndrome, and the NAFLD. A tumor suppressor PTEN negatively regulates the PI3K/AKT pathways through its lipid phosphatase activity. Molecular studies in the NAFLD support a key role for PTEN in hepatic insulin sensitivity and the development of steatosis, steatohepatitis, and fibrosis. We review recent studies on the features of the PTEN and the PI3K/AKT pathway and discuss the protein functions in the signaling pathways involved in the NAFLD. The molecular mechanisms contributing to the diseases are the subject of considerable investigation, as a better understanding of the pathogenesis will lead to novel therapies for a condition.
Collapse
|
493
|
Abstract
Many secreted polypeptide regulators of angiogenesis are devoid of signal peptides. These proteins are released through nonclassical pathways independent of endoplasmic reticulum and Golgi. In most cases, the nonclassical protein export is induced by stress. It usually serves to stimulate repair or inflammation in damaged tissues. We review the secreted signal peptide-less regulators of angiogenesis and discuss the mechanisms and biological significance of their unconventional export.
Collapse
Affiliation(s)
- Igor Prudovsky
- Maine Medical Center Research Institute, 81 Research Drive, Scarborough, ME 04074, USA
| |
Collapse
|
494
|
Kitagishi Y, Kobayashi M, Kikuta K, Matsuda S. Roles of PI3K/AKT/GSK3/mTOR Pathway in Cell Signaling of Mental Illnesses. DEPRESSION RESEARCH AND TREATMENT 2012; 2012:752563. [PMID: 23320155 PMCID: PMC3535741 DOI: 10.1155/2012/752563] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Revised: 11/09/2012] [Accepted: 11/21/2012] [Indexed: 12/26/2022]
Abstract
Several pharmacological agents acting on monoamine neurotransmission are used for the management of mental illnesses. Regulation of PI3K/AKT and GSK3 pathways may constitute an important signaling center in the subcellular integration of the synaptic neurotransmission. The pathways also modulate neuronal cell proliferation, migration, and plasticity. There are evidences to suggest that inflammation of neuron contributes to the pathology of depression. Inflammatory activation of neuron contributes to the loss of glial elements, which are consistent with pathological findings characterizing the depression. A mechanism of anti-inflammatory reactions from antidepressant medications has been found to be associated with an enhancement of heme oxygenase-1 expression. This induction in brain is also important in neuroprotection and neuroplasticity. As enzymes involved in cell survival and neuroplasticity are relevant to neurotrophic factor dysregulation, the PI3K/AKT/GSK3 may provide an important signaling for the neuroprotection in depression. In this paper, we summarize advances on the involvement of the PI3K/AKT/GSK3 pathways in cell signaling of neuronal cells in mental illnesses.
Collapse
Affiliation(s)
- Yasuko Kitagishi
- Department of Environmental Health Science, Nara Women's University, Kita-Uoya Nishimachi, Nara 630-8506, Japan
| | - Mayumi Kobayashi
- Department of Environmental Health Science, Nara Women's University, Kita-Uoya Nishimachi, Nara 630-8506, Japan
| | - Kanae Kikuta
- Department of Food Science and Nutrition, Nara Women's University, Nara 630-8506, Japan
| | - Satoru Matsuda
- Department of Environmental Health Science, Nara Women's University, Kita-Uoya Nishimachi, Nara 630-8506, Japan
| |
Collapse
|
495
|
Lin YM, Huang YL, Fong YC, Tsai CH, Chou MC, Tang CH. Hepatocyte growth factor increases vascular endothelial growth factor-A production in human synovial fibroblasts through c-Met receptor pathway. PLoS One 2012; 7:e50924. [PMID: 23209838 PMCID: PMC3508989 DOI: 10.1371/journal.pone.0050924] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2012] [Accepted: 10/26/2012] [Indexed: 11/19/2022] Open
Abstract
Background Angiogenesis is essential for the progression of osteoarthritis (OA). Hepatocyte growth factor (HGF) is an angiogenic mediator, and it shows elevated levels in regions of OA. However, the relationship between HGF and vascular endothelial growth factor (VEGF-A) in OA synovial fibroblasts (OASFs) is mostly unknown. Methodology/Principal Findings Here we found that stimulation of OASFs with HGF induced concentration- and time-dependent increases in VEGF-A expression. Pretreatment with PI3K inhibitor (Ly294002), Akt inhibitor, or mTORC1 inhibitor (rapamycin) blocked the HGF-induced VEGF-A production. Treatment of cells with HGF also increased PI3K, Akt, and mTORC1 phosphorylation. Furthermore, HGF increased the stability and activity of HIF-1 protein. Moreover, the use of pharmacological inhibitors or genetic inhibition revealed that c-Met, PI3K, Akt, and mTORC1 signaling pathways were potentially required for HGF-induced HIF-1α activation. Conclusions/Significance Taken together, our results provide evidence that HGF enhances VEGF-A expression in OASFs by an HIF-1α-dependent mechanism involving the activation of c-Met/PI3K/Akt and mTORC1 pathways.
Collapse
Affiliation(s)
- Yu-Min Lin
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Orthopedic Surgery, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Yuan-Li Huang
- Department of Biotechnology, College of Health Science, Asia University, Taichung, Taiwan
| | - Yi-Chin Fong
- Department of Orthopaedic Surgery, China Medical University Hospital, Taichung, Taiwan
- School of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Chun-Hao Tsai
- Department of Orthopaedic Surgery, China Medical University Hospital, Taichung, Taiwan
- Department of Medicine and Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan
| | - Ming-Chih Chou
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Chih-Hsin Tang
- Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
- * E-mail:
| |
Collapse
|
496
|
Copper-containing mesoporous bioactive glass scaffolds with multifunctional properties of angiogenesis capacity, osteostimulation and antibacterial activity. Biomaterials 2012; 34:422-33. [PMID: 23083929 DOI: 10.1016/j.biomaterials.2012.09.066] [Citation(s) in RCA: 477] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2012] [Accepted: 09/26/2012] [Indexed: 01/24/2023]
Abstract
It is of great importance to develop multifunctional bioactive scaffolds, which combine angiogenesis capacity, osteostimulation, and antibacterial properties for regenerating lost bone tissues. In order to achieve this aim, we prepared copper (Cu)-containing mesoporous bioactive glass (Cu-MBG) scaffolds with interconnective large pores (several hundred micrometer) and well-ordered mesopore channels (around 5 nm). Both Cu-MBG scaffolds and their ionic extracts could stimulate hypoxia-inducible factor (HIF)-1α and vascular endothelial growth factor (VEGF) expression in human bone marrow stromal cells (hBMSCs). In addition, both Cu-MBG scaffolds and their ionic extracts significantly promoted the osteogenic differentiation of hBMSCs by improving their bone-related gene expression (alkaline phosphatase (ALP), osteopontin (OPN) and osteocalcin (OCN)). Furthermore, Cu-MBG scaffolds could maintain a sustained release of ibuprofen and significantly inhibited the viability of bacteria. This study indicates that the incorporation of Cu(2+) ions into MBG scaffolds significantly enhances hypoxia-like tissue reaction leading to the coupling of angiogenesis and osteogenesis. Cu(2+) ions play an important role to offer the multifunctional properties of MBG scaffold system. This study has demonstrated that it is possible to develop multifunctional scaffolds by combining enhanced angiogenesis potential, osteostimulation, and antibacterial properties for the treatment of large bone defects.
Collapse
|