1
|
Zhuang Y, Liu K, He Q, Gu X, Jiang C, Wu J. Hypoxia signaling in cancer: Implications for therapeutic interventions. MedComm (Beijing) 2023; 4:e203. [PMID: 36703877 PMCID: PMC9870816 DOI: 10.1002/mco2.203] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 12/14/2022] [Accepted: 12/18/2022] [Indexed: 01/25/2023] Open
Abstract
Hypoxia is a persistent physiological feature of many different solid tumors and a key driver of malignancy, and in recent years, it has been recognized as an important target for cancer therapy. Hypoxia occurs in the majority of solid tumors due to a poor vascular oxygen supply that is not sufficient to meet the needs of rapidly proliferating cancer cells. A hypoxic tumor microenvironment (TME) can reduce the effectiveness of other tumor therapies, such as radiotherapy, chemotherapy, and immunotherapy. In this review, we discuss the critical role of hypoxia in tumor development, including tumor metabolism, tumor immunity, and tumor angiogenesis. The treatment methods for hypoxic TME are summarized, including hypoxia-targeted therapy and improving oxygenation by alleviating tumor hypoxia itself. Hyperoxia therapy can be used to improve tissue oxygen partial pressure and relieve tumor hypoxia. We focus on the underlying mechanisms of hyperoxia and their impact on current cancer therapies and discuss the prospects of hyperoxia therapy in cancer treatment.
Collapse
Affiliation(s)
- Yan Zhuang
- State Key Laboratory of Pharmaceutical BiotechnologyNational Institute of Healthcare Data Science at Nanjing UniversityJiangsu Key Laboratory of Molecular MedicineMedicineMedical School of Nanjing UniversityNanjing UniversityNanjingChina
| | - Kua Liu
- State Key Laboratory of Pharmaceutical BiotechnologyNational Institute of Healthcare Data Science at Nanjing UniversityJiangsu Key Laboratory of Molecular MedicineMedicineMedical School of Nanjing UniversityNanjing UniversityNanjingChina
| | - Qinyu He
- State Key Laboratory of Pharmaceutical BiotechnologyNational Institute of Healthcare Data Science at Nanjing UniversityJiangsu Key Laboratory of Molecular MedicineMedicineMedical School of Nanjing UniversityNanjing UniversityNanjingChina
| | - Xiaosong Gu
- Microecological, Regenerative and Microfabrication Technical Platform for Biomedicine and Tissue EngineeringJinan Microecological Biomedicine Shandong LaboratoryJinan CityChina
| | - Chunping Jiang
- State Key Laboratory of Pharmaceutical BiotechnologyNational Institute of Healthcare Data Science at Nanjing UniversityJiangsu Key Laboratory of Molecular MedicineMedicineMedical School of Nanjing UniversityNanjing UniversityNanjingChina
- Microecological, Regenerative and Microfabrication Technical Platform for Biomedicine and Tissue EngineeringJinan Microecological Biomedicine Shandong LaboratoryJinan CityChina
| | - Junhua Wu
- State Key Laboratory of Pharmaceutical BiotechnologyNational Institute of Healthcare Data Science at Nanjing UniversityJiangsu Key Laboratory of Molecular MedicineMedicineMedical School of Nanjing UniversityNanjing UniversityNanjingChina
- Microecological, Regenerative and Microfabrication Technical Platform for Biomedicine and Tissue EngineeringJinan Microecological Biomedicine Shandong LaboratoryJinan CityChina
| |
Collapse
|
2
|
Abstract
Gliomas are common brain mass with a high mortality rate. Patients with gliomas have a severely bad outcome, with an average survive duration less 15 months because of high recurrent rate and being resistant to radio-therapy and chemistry drugs therapy. Hyperbaric oxygen is extensively taken as an adjuvant treatment for various disease conditions. To know the characteristics of hyperbaric oxygen as a remedy for gliomas, we find that, in general, hyperbaric oxygen shows an obviously positive effect on the treatment of gliomas, and it can also relieve the complications caused by postoperative radiotherapy and chemotherapy of gliomas. Whereas, several researches have shown that hyperbaric oxygen promotes glioma progression.
Collapse
Affiliation(s)
- Wen-Jie Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Jia-Sheng Ding
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Qing Sun
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Xiang Xu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| |
Collapse
|
3
|
Lin SS, Niu CC, Yuan LJ, Tsai TT, Lai PL, Chong KY, Wei KC, Huang CY, Lu ML, Yang CY, Ueng SWN. Mir-573 regulates cell proliferation and apoptosis by targeting Bax in human degenerative disc cells following hyperbaric oxygen treatment. J Orthop Surg Res 2021; 16:16. [PMID: 33413477 PMCID: PMC7789655 DOI: 10.1186/s13018-020-02114-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 11/24/2020] [Indexed: 12/13/2022] Open
Abstract
Background MicroRNA (miRNA) plays a vital role in the intervertebral disc (IVD) degeneration. The expression level of miR-573 was downregulated whereas Bax was upregulated notably in human degenerative nucleus pulposus cells. In this study, we aimed to investigate the role of miR-573 in human degenerative nucleus pulposus (NP) cells following hyperbaric oxygen (HBO) treatment. Methods NP cells were separated from human degenerated IVD tissues. The control cells were maintained in 5% CO2/95% air and the hyperoxic cells were exposed to 100% O2 at 2.5 atmospheres absolute. MiRNA expression profiling was performed via microarray and confirmed by real-time PCR, and miRNA target genes were identified using bioinformatics and luciferase reporter assays. The mRNA and protein levels of Bax were measured. The proliferation of NPCs was detected using MTT assay. The protein expression levels of Bax, cleaved caspase 9, cleaved caspase 3, pro-caspase 9, and pro-caspase 3 were examined. Results Bioinformatics analysis indicated that the 3′ untranslated region (UTR) of the Bax mRNA contained the “seed-matched-sequence” for hsa-miR-573, which was validated via reporter assays. MiR-573 was induced by HBO and simultaneous suppression of Bax was observed in NP cells. Knockdown of miR-573 resulted in upregulation of Bax expression in HBO-treated cells. In addition, overexpression of miR-573 by HBO increased cell proliferation and coupled with inhibition of cell apoptosis. The cleavage of pro-caspase 9 and pro-caspase 3 was suppressed while the levels of cleaved caspase 9 and caspase 3 were decreased in HBO-treated cells. Transfection with anti-miR-573 partly suppressed the effects of HBO. Conclusion Mir-573 regulates cell proliferation and apoptosis by targeting Bax in human degenerative NP cells following HBO treatment.
Collapse
Affiliation(s)
- Song-Shu Lin
- Department of Orthopaedic Surgery, Chang Gung Memorial Hospital, No 5, Fu-Hsing Street, Linkou, Taoyuan, 333, Taiwan.,Department of Nursing, Chang Gung University of Science and Technology, Taoyuan, Taiwan.,Hyperbaric Oxygen Medical Research Lab, Bone and Joint Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Chi-Chien Niu
- Department of Orthopaedic Surgery, Chang Gung Memorial Hospital, No 5, Fu-Hsing Street, Linkou, Taoyuan, 333, Taiwan.,Hyperbaric Oxygen Medical Research Lab, Bone and Joint Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Li-Jen Yuan
- Department of Orthopaedic Surgery, E-Da Hospital/I-Shou University, Kaohsiung, Taiwan
| | - Tsung-Ting Tsai
- Department of Orthopaedic Surgery, Chang Gung Memorial Hospital, No 5, Fu-Hsing Street, Linkou, Taoyuan, 333, Taiwan.,Hyperbaric Oxygen Medical Research Lab, Bone and Joint Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Po-Liang Lai
- Department of Orthopaedic Surgery, Chang Gung Memorial Hospital, No 5, Fu-Hsing Street, Linkou, Taoyuan, 333, Taiwan.,Hyperbaric Oxygen Medical Research Lab, Bone and Joint Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Kowit-Yu Chong
- Hyperbaric Oxygen Medical Research Lab, Bone and Joint Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan.,Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan
| | - Kuo-Chen Wei
- Hyperbaric Oxygen Medical Research Lab, Bone and Joint Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan.,Department of Neurosugery, New Taipei Municipal Tu Cheng Hospital, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
| | - Chiung-Yin Huang
- Hyperbaric Oxygen Medical Research Lab, Bone and Joint Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan.,Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan
| | - Meng-Ling Lu
- Department of Orthopaedic Surgery, Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Chuen-Yung Yang
- Department of Orthopaedic Surgery, Chang Gung Memorial Hospital, No 5, Fu-Hsing Street, Linkou, Taoyuan, 333, Taiwan.,Hyperbaric Oxygen Medical Research Lab, Bone and Joint Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Steve W N Ueng
- Department of Orthopaedic Surgery, Chang Gung Memorial Hospital, No 5, Fu-Hsing Street, Linkou, Taoyuan, 333, Taiwan. .,Hyperbaric Oxygen Medical Research Lab, Bone and Joint Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan. .,College of Medicine, Chang Gung University, Taoyuan, Taiwan.
| |
Collapse
|
4
|
Song K, Chen J, Ding J, Xu H, Xu H, Qin Z. Hyperbaric oxygen suppresses stemness-associated properties and Nanog and oncostatin M expression, but upregulates β-catenin in orthotopic glioma models. J Int Med Res 2019; 48:300060519872898. [PMID: 31813325 PMCID: PMC7607208 DOI: 10.1177/0300060519872898] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Objective This study aimed to explore whether initial hyperbaric oxygen treatment
affects the stemness of glioma stem cells using an in vivo
basal ganglia glioma model. Methods A basal ganglia glioma rat model was established. Rats were exposed to normal
oxygen or hyperbaric oxygen on days 2, 4, 6, 8, 10, and 12. After 16 days of
glioma cell inoculation, western blot, ELISA, and flow cytometry were
performed to examine stemness-associated properties by examining the
expression of CD133, A2B5, Nanog, oncostatin M, β-catenin, Oct-3/4, Sox2,
and Nestin. Results Initial hyperbaric oxygen treatment began to affect glioma
stemness-associated properties. The proportion of
CD133+A2B5+ cells was significantly reduced after
initial hyperbaric oxygen treatment. Additionally, the expression of
stemness-related genes such as Nanog and oncostatin M was reduced, while
TGF-β and β-catenin were increased. Conclusions Initial hyperbaric oxygen treatment not only alters the hypoxic
microenvironment but also affects the stemness-associated properties of
cancer stem cells.
Collapse
Affiliation(s)
- Kun Song
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Junrui Chen
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jianbo Ding
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hao Xu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hongzhi Xu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhiyong Qin
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
5
|
Zembrzuska K, Ostrowski RP, Matyja E. Hyperbaric oxygen increases glioma cell sensitivity to antitumor treatment with a novel isothiourea derivative in vitro. Oncol Rep 2019; 41:2703-2716. [PMID: 30896865 PMCID: PMC6448092 DOI: 10.3892/or.2019.7064] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 03/04/2019] [Indexed: 12/27/2022] Open
Abstract
Glioblastoma (GBM) is the most common primary brain tumor. Tumor hypoxia is a pivotal factor responsible for the progression of this malignant glioma, and its resistance to radiation and chemotherapy. Thus, improved tumor tissue oxygenation may promote greater sensitivity to anticancer treatment. Protein kinase D1 (PKD1) protects cells from oxidative stress, and its abnormal activity serves an important role in multiple malignancies. The present study examined the effects of various oxygen conditions on the cytotoxic potential of the novel isothiourea derivate N,N′-dimethyl-S-(2,3,4,5,6-pentabromobenzyl)- isothiouronium bromide (ZKK-3) against the T98G GBM cell line. ZKK-3 was applied at concentrations of 10, 25 and 50 µM, and cells were maintained under conditions of normoxia, anoxia, hypoxia, hyperbaric oxygen (HBO), hypoxia/hypoxia and hypoxia/HBO. The proliferation and viability of neoplastic cells, and protein expression levels of hypoxia-inducible factor 1α (HIF-1α), PKD1, phosphorylated (p)PKD1 (Ser 916) and pPKD1 (Ser 744/748) kinases were evaluated. Oxygen deficiency, particularly regarding hypoxia, could diminish the cytotoxic effect of ZKK-3 at 25 and 50 µM and improve T98G cell survival compared with normoxia. HBO significantly reduced cell proliferation and increased T98G cell sensitivity to ZKK-3 when compared with normoxia. HIF-1α expression levels were increased under hypoxia compared with normoxia and decreased under HBO compared with hypoxia/hypoxia at 0, 10 and 50 µM ZKK-3, suggesting that HBO improved oxygenation of the cells. ZKK-3 exhibited inhibitory activity against pPKD1 (Ser 916) kinase; however, the examined oxygen conditions did not appear to significantly influence the expression of this phosphorylated form in cells treated with the tested compound. Regarding pPKD1 (Ser 744/748), a significant difference in expression was observed only for cells treated with 10 µM ZKK-3 and hypoxia/hypoxia compared with normoxia. However, there were significant differences in the expression levels of both phosphorylated forms of PKD1 under different oxygen conditions in the controls. In conclusion, the combination of isothiourea derivatives and hyperbaric oxygenation appears to be a promising therapeutic approach for malignant glioma treatment.
Collapse
Affiliation(s)
- Katarzyna Zembrzuska
- Department of Experimental and Clinical Neuropathology, Mossakowski Medical Research Centre, Polish Academy of Sciences, 02‑106 Warsaw, Poland
| | - Robert P Ostrowski
- Department of Experimental and Clinical Neuropathology, Mossakowski Medical Research Centre, Polish Academy of Sciences, 02‑106 Warsaw, Poland
| | - Ewa Matyja
- Department of Experimental and Clinical Neuropathology, Mossakowski Medical Research Centre, Polish Academy of Sciences, 02‑106 Warsaw, Poland
| |
Collapse
|
6
|
miR-1268a regulates ABCC1 expression to mediate temozolomide resistance in glioblastoma. J Neurooncol 2018; 138:499-508. [DOI: 10.1007/s11060-018-2835-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 02/22/2018] [Indexed: 02/07/2023]
|
7
|
Wang YG, Long J, Shao DC, Song H. Hyperbaric oxygen inhibits production of CD3+ T cells in the thymus and facilitates malignant glioma cell growth. J Int Med Res 2018; 46:2780-2791. [PMID: 29785863 PMCID: PMC6124287 DOI: 10.1177/0300060518767796] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Objective Hyperbaric oxygen (HBO) is an emerging complementary alternative medical approach in glioma treatment. However, its mode of action is unknown, so this was investigated in the present study. Methods We constructed an intracranial glioma model of congenic C57BL/6J mice. Glioma growth under HBO stimulation was assessed by bioluminescent imaging and magnetic resonance imaging. Flow cytometry assessed direct effects of HBO on reactive oxygen species (ROS) signaling of transplanted glioma cells and organs, and quantified mature T cells and subgroups in tumors, the brain, and blood. Results HBO promoted the growth of transplanted GL261-Luc glioma in the intracranial glioma mouse model. ROS signaling of glioma cells and brain cells was significantly downregulated under HBO stimulation, but thymus ROS levels were significantly upregulated. CD3+ T cells were significantly downregulated, while both Ti/Th cells (CD3+CD4+) and Ts/Tc cells (CD3+CD8+) were inhibited in tumors of the HBO group. The percentage of regulatory T cells in Ti/Th (CD3+CD4+) cells was elevated in the tumors and thymuses of the HBO group. Conclusion HBO induced ROS signaling in the thymus, inhibited CD3+ T cell generation, and facilitated malignant glioma cell growth in vivo in the intracranial glioma mouse model.
Collapse
Affiliation(s)
- Yong-Gang Wang
- 1 Department of Neurosurgery, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
| | - Jiang Long
- 1 Department of Neurosurgery, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
| | - Dong-Chuan Shao
- 2 Department of Neurosurgery, First People's Hospital of Kunming, Kunming, Yunnan 650032, China
| | - Hai Song
- 1 Department of Neurosurgery, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
| |
Collapse
|
8
|
Huang L, Boling W, Zhang JH. Hyperbaric oxygen therapy as adjunctive strategy in treatment of glioblastoma multiforme. Med Gas Res 2018; 8:24-28. [PMID: 29770193 PMCID: PMC5937300 DOI: 10.4103/2045-9912.229600] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most common type of malignant intracranial tumor in adults. Tumor tissue hypoxia, high mitotic rate, and rapid tumor spread account for its poor prognosis. Hyperbaric oxygen therapy (HBOT) may improve the sensitivity of radio-chemotherapy by increasing oxygen tension within the hypoxic regions of the neoplastic tissue. This review summarizes the research of HBOT applications within the context of experimental and clinical GBM. Limited clinical trials and preclinical studies suggest that radiotherapy immediately after HBOT enhances the effects of radiotherapy in some aspects. HBOT also is able to strengthen the anti-tumor effect of chemotherapy when applied together. Overall, HBOT is well tolerated in the GBM patients and does not significantly increase toxicity. However, HBOT applied by itself as curative strategy against GBM is controversial in preclinical studies and has not been evaluated rigorously in GBM patients. In addition to HBOT favorably managing the therapeutic resistance of GBM, future research needs to focus on the multimodal or cocktail approaches to treatment, as well as molecular strategies targeting GBM stem cells.
Collapse
Affiliation(s)
- Lei Huang
- Department of Neurosurgery, School of Medicine, Loma Linda University, Loma Linda, CA, USA.,Department of Basic Sciences, Division of Physiology, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Warren Boling
- Department of Neurosurgery, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - John H Zhang
- Department of Neurosurgery, School of Medicine, Loma Linda University, Loma Linda, CA, USA.,Department of Basic Sciences, Division of Physiology, School of Medicine, Loma Linda University, Loma Linda, CA, USA.,Department of Anesthesiology, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| |
Collapse
|
9
|
Xie Y, Zeng X, Wu X, Hu J, Zhu Y, Yang X. Hyperbaric oxygen as an adjuvant to temozolomide nanoparticle inhibits glioma growth by inducing G2/M phase arrest. Nanomedicine (Lond) 2018; 13:887-898. [PMID: 29473458 DOI: 10.2217/nnm-2017-0395] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
AIM To study the effects of combinational treatment of hyperbaric oxygen (HBO) and nanotemozolomide in glioma. MATERIALS & METHODS Temozolomide (TMZ)-loaded porous silicon nanoparticles (TMZ/PSi NPs) were prepared. In vitro and in vivo evaluations were performed. RESULTS The cell uptake of TMZ/PSi NPs could be tracked by autofluorescence of porous silicon. The concentration of oxygen in tumor was improved and the antitumor rate was increased to 84.2% in the TMZ/PSi NPs combined with HBO group. The viability of hypoxia-induced glioma C6 cells was decreased and cell cycle was arrested at G2/M phase in response to TMZ/PSi NPs treatment with HBO compared with continuous treatment with hypoxia. CONCLUSION The combinational treatment of TMZ/PSi NPs and HBO could be a promising therapeutic strategy for glioma.
Collapse
Affiliation(s)
- Yuanyuan Xie
- College of Life Science & Technology, National Engineering Research Center for Nanomedicine, Huazhong University of Science & Technology, Wuhan, 430074, PR China
| | - Xiaofan Zeng
- College of Life Science & Technology, National Engineering Research Center for Nanomedicine, Huazhong University of Science & Technology, Wuhan, 430074, PR China
| | - Xian Wu
- College of Life Science & Technology, National Engineering Research Center for Nanomedicine, Huazhong University of Science & Technology, Wuhan, 430074, PR China
| | - Jun Hu
- College of Life Science & Technology, National Engineering Research Center for Nanomedicine, Huazhong University of Science & Technology, Wuhan, 430074, PR China
| | - Yanhong Zhu
- College of Life Science & Technology, National Engineering Research Center for Nanomedicine, Huazhong University of Science & Technology, Wuhan, 430074, PR China
| | - Xiangliang Yang
- College of Life Science & Technology, National Engineering Research Center for Nanomedicine, Huazhong University of Science & Technology, Wuhan, 430074, PR China
| |
Collapse
|
10
|
HIF1α regulates glioma chemosensitivity through the transformation between differentiation and dedifferentiation in various oxygen levels. Sci Rep 2017; 7:7965. [PMID: 28801626 PMCID: PMC5554160 DOI: 10.1038/s41598-017-06086-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 06/07/2017] [Indexed: 12/20/2022] Open
Abstract
Chemotherapy plays a significant role in glioma treatment; however, it has limited effectiveness in extending the life expectancies of glioma patients. Traditional studies have attributed this lack of efficacy to glioma stem cells (GSCs) and their high resistance to chemotherapy, and hypoxia worsens this issue. In contrast, hyperoxia effectively alleviates hypoxia in glioma and sensitizes glioma cells to chemotherapy. In a summary of traditional studies, the majority of researchers overlooked the influence of hypoxia on differentiated cells because they only focused on the maintenance of GSCs stemness, which thus resulted in chemoresistance. Because of this background, we hypothesized that GSCs may be induced through dedifferentiation under hypoxic conditions, and hypoxia maintains GSCs stemness, which thus leads to resistance to chemotherapy. In contrast, hyperoxia inhibits the dedifferentiation process and promotes GSCs differentiation, which increases the sensitization of glioma cells to chemotherapy. Hypoxia-inducible factor-1α (HIF1α) contributes substantially to the stemness maintenance of GSCs and resistance of glioma to chemotherapy; thus, we investigated whether HIF1α regulates the resistance or sensitization of glioma cells to chemotherapy in different oxygen levels. It highlights a novel viewpoint on glioma chemosensitivity from the transformation between dedifferentiation and differentiation in different oxygen levels.
Collapse
|
11
|
Qi Y, Ruan J, Wang M, Dai Y, Zhou Q, Gui S, Zhang S, Wang Y. Effects of hyperbaric oxygen treatment on gastric cancer cell line SGC7901. Biomed Rep 2017; 6:475-479. [PMID: 28413648 DOI: 10.3892/br.2017.869] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 02/14/2017] [Indexed: 01/02/2023] Open
Abstract
Hyperbaric oxygen (HBO) has been previously identified as an effective adjunct treatment option for the management of brain injury, diabetic ulcers and chronic wounds. However, the roles of HBO as an adjunctive therapy for tumors remain controversial. The present research project was performed to explore the effects of HBO treatment on proliferation, autophagy and endoplasmic reticulum stress response of the gastric cancer cell line, SGC7901. The present study demonstrated that, after subjecting SGC7901 cells to HBO treatment, the increase in cell proliferation was significant, compared with that of the control group. In addition, there was a significant increase in LC3-phosphatidylethanolamine conjugate (LC3-II) level, as well as binding immunoglobulin protein level, and a significant decrease in CCAAT-enhancer-binding protein homologous protein level. These suggested that hyperbaric oxygen treatment alone may promote proliferation and cell survival of gastric cancer cell SGC7901, and inhibit apoptosis through regulating cell autophagy and oxidative stress.
Collapse
Affiliation(s)
- Yinliang Qi
- General Department of Hyperbaric Oxygen, The Second People's Hospital of Hefei, Hefei, Anhui 230011, P.R. China
| | - Jianfeng Ruan
- General Department of Hyperbaric Oxygen, The Second People's Hospital of Hefei, Hefei, Anhui 230011, P.R. China
| | - Mei Wang
- General Department of Hyperbaric Oxygen, The Second People's Hospital of Hefei, Hefei, Anhui 230011, P.R. China
| | - Yuanchang Dai
- General Department of Hyperbaric Oxygen, The Second People's Hospital of Hefei, Hefei, Anhui 230011, P.R. China
| | - Qing Zhou
- Laboratory of Molecular Biology and Department of Biochemistry, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Shuyu Gui
- Department of Respiratory Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230011, P.R. China
| | - Sumei Zhang
- Laboratory of Molecular Biology and Department of Biochemistry, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Yuan Wang
- Laboratory of Molecular Biology and Department of Biochemistry, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| |
Collapse
|
12
|
Ding JB, Chen JR, Xu HZ, Qin ZY. Effect of Hyperbaric Oxygen on the Growth of Intracranial Glioma in Rats. Chin Med J (Engl) 2016; 128:3197-203. [PMID: 26612296 PMCID: PMC4794883 DOI: 10.4103/0366-6999.170278] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Background: Numerous studies have confirmed that hyperbaric oxygen (HBO) in combination with radiotherapy or chemotherapy may increase the efficacy of radiotherapy or chemotherapy in patients with glioma. However, whether HBO therapy alone may inhibit or promote the growth of malignant tumors remains controversial. This study aimed to investigate the effect of HBO on the growth of glioma in rats, and the impact of HBO on the expression of vascular endothelial growth factor (VEGF) and hypoxia-inducible factor 1-alpha (HIF-1α), angiogenesis, and apoptosis of glioma cells. Methods: Male Sprague–Dawley rats were treated with or without HBO after glioma cell inoculation and followed for up to 16 days postinoculation. Rats were randomized to receive bilateral forelimb function tests (n = 20 per group) and head magnetic resonance imaging (n = 5 per group). Differences between HBO and control groups were tested using 2-sample independent t-tests and changes over time within treatment groups were analyzed using a repeated measurement analysis of variance with Bonferroni correction. The effect of HBO on the expression of VEGF, HIF-1α, von Willebrand factor, angiogenesis, and tumor cell apoptosis were also examined (n = 5 per group). Results: Forelimb function scores were reduced in both HBO-treated and control groups. HBO-treated rats had significantly larger tumor volume and more water in the cerebellum compared with control rats. The intratumoral expression of VEGF was significantly higher in HBO-treated rats compared with control rats (23.2% vs. 13.3%, P = 0.002). HIF-1α was significantly increased in HBO-treated rats compared with controls in the expression of both intratumoral (72.7% vs. 54.9%, P = 0.001) and peritumoral (2.6% vs. 1.9%, P = 0.003) cells. The intratumoral microvessel density (MVD) was significantly higher in the HBO group (15.6 vessels/field vs. 4.4 vessels/field, P < 0.001), and the peritumoral MVD was not significantly different between the two groups (P > 0.05). Apoptosis was significantly lower in HBO-treated rats compared with controls (44.4% vs. 82.8% for intratumoral; 10.1% vs. 77.5% for peritumoral, both P < 0.001). Conclusions: The current results demonstrate that HBO alone may promote tumor growth, and is therefore not suitable to treat patients with gliomas with neurological deficits or disorders with HBO alone. If HBO must be used as a mean of rehabilitation, it is recommended that HBO should be combined with radiotherapy or chemotherapy.
Collapse
Affiliation(s)
| | | | | | - Zhi-Yong Qin
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China
| |
Collapse
|
13
|
Stępień K, Ostrowski RP, Matyja E. Hyperbaric oxygen as an adjunctive therapy in treatment of malignancies, including brain tumours. Med Oncol 2016; 33:101. [PMID: 27485098 PMCID: PMC4971045 DOI: 10.1007/s12032-016-0814-0] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 07/16/2016] [Indexed: 12/22/2022]
Abstract
Hyperbaric oxygen (HBO) therapy is widely used as an adjunctive treatment for various pathological states, predominantly related to hypoxic and/or ischaemic conditions. It also holds promise as an approach to overcoming the problem of oxygen deficiency in the poorly oxygenated regions of the neoplastic tissue. Occurrence of local hypoxia within the central areas of solid tumours is one of the major issues contributing to ineffective medical treatment. However, in anti-cancer therapy, HBO alone gives a limited curative effect and is typically not applied by itself. More often, HBO is used as an adjuvant treatment along with other therapeutic modalities, such as radio- and chemotherapy. This review outlines the existing data regarding the medical use of HBO in cancer treatment, with a particular focus on the use of HBO in the treatment of brain tumours. We conclude that the administration of HBO can provide many clinical benefits in the treatment of tumours, including management of highly malignant gliomas. Applied immediately before irradiation, it is safe and well tolerated by patients, causing rare and limited side effects. The results obtained with a combination of HBO/radiotherapy protocol proved to be especially favourable compared to radiation treatment alone. HBO can also increase the cytostatic effect of certain drugs, which may render standard chemotherapy more effective. The currently available data support the legitimacy of conducting further research on the use of HBO in the treatment of malignancies.
Collapse
Affiliation(s)
- Katarzyna Stępień
- Department of Experimental and Clinical Neuropathology, Mossakowski Medical Research Centre, Polish Academy of Sciences, 5 Pawińskiego Str., 02-106, Warsaw, Poland.
| | - Robert P Ostrowski
- Department of Experimental and Clinical Neuropathology, Mossakowski Medical Research Centre, Polish Academy of Sciences, 5 Pawińskiego Str., 02-106, Warsaw, Poland
| | - Ewa Matyja
- Department of Experimental and Clinical Neuropathology, Mossakowski Medical Research Centre, Polish Academy of Sciences, 5 Pawińskiego Str., 02-106, Warsaw, Poland
| |
Collapse
|
14
|
Sun C, Yu Y, Wang L, Wu B, Xia L, Feng F, Ling Z, Wang S. Additive antiangiogenesis effect of ginsenoside Rg3 with low-dose metronomic temozolomide on rat glioma cells both in vivo and in vitro. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2016; 35:32. [PMID: 26872471 PMCID: PMC4752767 DOI: 10.1186/s13046-015-0274-y] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 12/17/2015] [Indexed: 11/23/2022]
Abstract
Background Glioblastoma is the most common and deadly primary brain tumor in adults. Low-dose,metronomic (LDM) temozolomide (TMZ) displays improved efficacy in the treatment of glioblastoma by targeting angiogenesis, but has a limited effect on recurrence. The antiangiogenesis drug ginsenoside Rg3 (RG3) is the main active ingredient of ginseng, a popular herbal medicine. Methods Using an in vitro and a rat model of an orthotopic glioma allograft, this study was to determine whether RG3 enhanced the antiangiogenesis activity of LDM TMZ in the treatment of glioblastoma. Results Our results showed that combined use of TMZ with RG3 displayed additive inhibition on proliferation of both human umbilical vein endothelial cells (HUVEC) and rat C6 glioma cells in vitro. They additively arrested cell cycle, increased apoptosis, and decreased VEGF-A and BCL-2 expression in HUVEC. Antiangiogenesis effect was also evaluated in the rat model of orthotopic glioma allograft, based upon markers including relative cerebral blood volume (rCBV) by magnetic resonance imaging (MRI), VEGF levels and microvessel density (MVD)/CD34 staining. LDM TMZ alone was potent in suppressing angiogenesis and tumor growth, whereas RG3 alone only had modest antiangiogenesis effects. Combined treatment significantly and additively suppressed angiogenesis, without additive inhibitory effects on allografted tumor growth. Conclusions These data provide evidence showing the efficacy of LDM TMZ on glioma treatment. The combined additive antiangiogenesis effect suggests that RG3 has the potential to further increase the efficacy of LDM TMZ in the treatment of glioblastoma. Electronic supplementary material The online version of this article (doi:10.1186/s13046-015-0274-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Caixing Sun
- Department of Neurosurgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, 310022, China.
| | - Yang Yu
- Department of Neurosurgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, 310022, China.
| | - Lizhen Wang
- Department of Neurosurgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, 310022, China.
| | - Bin Wu
- Department of Neurosurgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, 310022, China.
| | - Liang Xia
- Department of Neurosurgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, 310022, China.
| | - Fang Feng
- Department of Neurosurgery, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, 310022, China.
| | - Zhiqiang Ling
- Zhejiang Cancer Research Institute, Zhejiang Cancer Hospital, 38 Guangji Road, Hangzhou, Zhejiang, 310022, China.
| | - Shihua Wang
- Department of Cancer Biology, Wake Forest School of Medicine, Medical Center Boulevard, NC, 27157, USA.
| |
Collapse
|
15
|
He H, Yao M, Zhang W, Tao B, Liu F, Li S, Dong Y, Zhang C, Meng Y, Li Y, Hu G, Luo C, Zong H, Lu Y. MEK2 is a prognostic marker and potential chemo-sensitizing target for glioma patients undergoing temozolomide treatment. Cell Mol Immunol 2015; 13:658-68. [PMID: 26189368 DOI: 10.1038/cmi.2015.46] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Revised: 04/30/2015] [Accepted: 05/01/2015] [Indexed: 02/06/2023] Open
Abstract
Although temozolomide (TMZ) is the first-line chemotherapeutic agent for glioblastoma, it is often non-curative due to drug resistance. To overcome the resistance of glioblastoma cells to TMZ, it is imperative to identify prognostic markers for outcome prediction and to develop chemo-sensitizing agents. Here, the gene expression profiles of TMZ-resistant and TMZ-sensitive samples were compared by microarray analysis, and mitogen-activated protein kinase kinase 2 (MEK2) was upregulated specifically in resistant glioma cells but not in sensitive tumor cells or non-tumor tissues. Moreover, a comprehensive analysis of patient data revealed that the increased level of MEK2 expression correlated well with the advancement of glioma grade and worse prognosis in response to TMZ treatment. Furthermore, reducing the level of MEK2 in U251 glioma cell lines or xenografted glioma models through shRNA-mediated gene knockdown inhibited cell proliferation and enhanced the sensitivity of cells toward TMZ treatment. Further analysis of tumor samples from glioma patients by real-time PCR indicated that an increased MEK2 expression level was closely associated with the activation of many drug resistance genes. Finally, these resistance genes were downregulated after MEK2 was silenced in vitro, suggesting that the mechanism of MEK2-induced chemo-resistance could be mediated by the transcriptional activation of these resistance genes. Collectively, our data indicated that the expression level of MEK2 could serve as a prognostic marker for glioma chemotherapy and that MEK2 antagonists can be used as chemo-sensitizers to enhance the treatment efficacy of TMZ.
Collapse
Affiliation(s)
- Hua He
- Department of Neurosurgery, Changzheng Hospital, Second Affiliated Hospital of Second Military Medical University, 415 Fengyang Road, Shanghai 200003, P.R.China
| | - Maojin Yao
- Department of Microbiology, Immunology and Cancer Biology, School of Medicine, University of Virginia, USA
| | - Wenhao Zhang
- Department of Hematology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Bangbao Tao
- Department of Neurosurgery, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Feili Liu
- Department of Neurosurgery, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Shu Li
- Department of Pathophysiology, Wannan Medical College, 22 Wenchang Road Wuhu 241002, China
| | - Yan Dong
- Department of Neurosurgery, Changzheng Hospital, Second Affiliated Hospital of Second Military Medical University, 415 Fengyang Road, Shanghai 200003, P.R.China
| | - Chenran Zhang
- Department of Neurosurgery, Changzheng Hospital, Second Affiliated Hospital of Second Military Medical University, 415 Fengyang Road, Shanghai 200003, P.R.China
| | - Yicheng Meng
- Department of Neurosurgery, Changzheng Hospital, Second Affiliated Hospital of Second Military Medical University, 415 Fengyang Road, Shanghai 200003, P.R.China
| | - Yuxin Li
- Department of Neurosurgery, Changzheng Hospital, Second Affiliated Hospital of Second Military Medical University, 415 Fengyang Road, Shanghai 200003, P.R.China
| | - Guohan Hu
- Department of Neurosurgery, Changzheng Hospital, Second Affiliated Hospital of Second Military Medical University, 415 Fengyang Road, Shanghai 200003, P.R.China
| | - Chun Luo
- Department of Neurosurgery, Changzheng Hospital, Second Affiliated Hospital of Second Military Medical University, 415 Fengyang Road, Shanghai 200003, P.R.China
| | - Hui Zong
- Department of Microbiology, Immunology and Cancer Biology, School of Medicine, University of Virginia, USA
| | - Yicheng Lu
- Department of Neurosurgery, Changzheng Hospital, Second Affiliated Hospital of Second Military Medical University, 415 Fengyang Road, Shanghai 200003, P.R.China
| |
Collapse
|
16
|
WANG YONGGANG, ZHAN YIPING, PAN SHUYI, WANG HAIDONG, ZHANG DUNXIAO, GAO KAI, QI XUELING, YU CHUNJIANG. Hyperbaric oxygen promotes malignant glioma cell growth and inhibits cell apoptosis. Oncol Lett 2015; 10:189-195. [PMID: 26170997 PMCID: PMC4487135 DOI: 10.3892/ol.2015.3244] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Accepted: 04/13/2015] [Indexed: 11/06/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most frequently diagnosed intracranial malignant tumor in adults. Clinical studies have indicated that hyperbaric oxygen may improve the prognosis and reduce complications in glioma patients; however, the specific mechanism by which this occurs remains unknown. The present study investigated the direct effects of hyperbaric oxygen stimulation on glioma by constructing an intracranial transplanted glioma model in congenic C57BL/6J mice. Bioluminescent imaging (BLI) was used to assess the growth of intracranial transplanted GL261-Luc glioma cells in vivo, while flow cytometric and immunohistochemical assays were used to detect and compare the expression of the biomarkers, Ki-67, CD34 and TUNEL, reflecting the cell cycle, apoptosis and angiogenesis. BLI demonstrated that hyperbaric oxygen promoted the growth of intracranially transplanted GL261-Luc glioma cells in vivo. Flow cytometric analysis indicated that hyperbaric oxygen promoted GL261-Luc glioma cell proliferation and also prevented cell cycle arrest. In addition, hyperbaric oxygen inhibited the apoptosis of the transplanted glioma cells. Immunohistochemical analysis also indicated that hyperbaric oxygen increased positive staining for Ki-67 and CD34, while reducing staining for TUNEL (a marker of apoptosis). The microvessel density was significantly increased in the hyperbaric oxygen treatment group compared with the control group. In conclusion, hyperbaric oxygen treatment promoted the growth of transplanted malignant glioma cells in vivo and also inhibited the apoptosis of these cells.
Collapse
Affiliation(s)
- YONG-GANG WANG
- Department of Neurosurgery, Beijing Sanbo Brain Hospital, Capital Medical University, Beijing 100093, P.R. China
| | - YI-PING ZHAN
- Department of Neurosurgery, Beijing Sanbo Brain Hospital, Capital Medical University, Beijing 100093, P.R. China
| | - SHU-YI PAN
- Department of Hyperbaric Oxygen, Navy General Hospital, Beijing 100048, P.R. China
| | - HAI-DONG WANG
- Department of Hyperbaric Oxygen, Navy General Hospital, Beijing 100048, P.R. China
| | - DUN-XIAO ZHANG
- Department of Hyperbaric Oxygen, Navy General Hospital, Beijing 100048, P.R. China
| | - KAI GAO
- Institute of Laboratory Animal Sciences, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing 100021, P.R. China
| | - XUE-LING QI
- Department of Pathology, Beijing Sanbo Brain Hospital, Capital Medical University, Beijing 100093, P.R. China
| | - CHUN-JIANG YU
- Department of Neurosurgery, Beijing Sanbo Brain Hospital, Capital Medical University, Beijing 100093, P.R. China
| |
Collapse
|
17
|
Montaldi AP, Godoy PRDV, Sakamoto-Hojo ET. APE1/REF-1 down-regulation enhances the cytotoxic effects of temozolomide in a resistant glioblastoma cell line. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2015; 793:19-29. [PMID: 26520369 DOI: 10.1016/j.mrgentox.2015.06.001] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2015] [Accepted: 06/02/2015] [Indexed: 01/25/2023]
Abstract
Temozolomide (TMZ) is widely used for patients with glioblastoma (GBM); however, tumor cells frequently exhibit drug-resistance. Base excision repair (BER) has been identified as a possible mediator of TMZ resistance, and an attractive approach to sensitizing cells to chemotherapy. Human apurinic/apyrimidinic endonuclease/redox factor-1 (APE1) is an essential enzyme with a role in the BER pathway by repairing abasic sites, and it also acts as a reduction factor, maintaining transcription factors in an active reduced state. Thus, we aimed to investigate whether the down-regulation of APE1 expression by siRNA can interfere with the resistance of GBM to TMZ, being evaluated by several cellular and molecular parameters. We demonstrated that APE1 knockdown associated with TMZ treatment efficiently reduced cell proliferation and clonogenic survival of resistant cells (T98G), which appears to be a consequence of increased DNA damage, S-phase arrest, and H2AX phosphorylation, resulting in apoptosis induction. On the contrary, for those assays, the sensitization effects of APE1 silencing plus TMZ treatment did not occur in the TMZ-sensitive cell line (U87MG). Interestingly, TMZ-treatment and APE1 knockdown significantly reduced cell invasion in both cell lines, but TMZ alone did not reduce the invasion capacity of U87MG cells, as observed for T98G. We also found that VEGF expression was down-regulated by TMZ treatment in T98G cells, regardless of APE1 knockdown, but U87MG showed a different response, since APE1 silencing counteracted VEGF induction promoted by TMZ, suggesting that the APE1-redox function may play an indirect role, depending on the cell line. The present results support the contribution of BER in the GBM resistance to TMZ, with a greater effect in TMZ-resistant, compared with TMZ-sensitive cells, emphasizing that APE1 can be a promising target for modifying TMZ tolerance. Furthermore, genetic characteristics of tumor cells should be considered as critical information to select an appropriate therapeutic strategy.
Collapse
Affiliation(s)
- Ana P Montaldi
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo (USP), Brazil; Department of Biology, Faculty of Philosophy, Sciences and Letters at Ribeirão Preto - University of São Paulo (USP), Ribeirão Preto, S.P., Brazil
| | - Paulo R D V Godoy
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo (USP), Brazil; Department of Biology, Faculty of Philosophy, Sciences and Letters at Ribeirão Preto - University of São Paulo (USP), Ribeirão Preto, S.P., Brazil
| | - Elza T Sakamoto-Hojo
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo (USP), Brazil; Department of Biology, Faculty of Philosophy, Sciences and Letters at Ribeirão Preto - University of São Paulo (USP), Ribeirão Preto, S.P., Brazil.
| |
Collapse
|
18
|
Ye X, Zhang J, Lu R, Zhou G. HBO: a possible supplementary therapy for oral potentially malignant disorders. Med Hypotheses 2014; 83:131-6. [PMID: 24908359 DOI: 10.1016/j.mehy.2014.05.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Revised: 04/15/2014] [Accepted: 05/13/2014] [Indexed: 12/25/2022]
Abstract
Oral potentially malignant disorders (OPMDs) are chronic inflammatory diseases in which cells suffer hypoxia referring to deprivation of adequate oxygen supply. Hyperbaric oxygen treatment (HBO), which can increase oxygen tension and delivery to oxygen-deficient tissue, is a supplementary therapy to improve or cure disorders involving hypoxia. Although the applications of HBO in wound healings, acute ischemic stroke, radiation-induced soft tissue injury and cancers are extensively reported, there are only few studies on their effect in OPMDs. Not only does HBO furnish oxygen-it also possesses potent anti-inflammatory properties. At the cellular level, HBO can decrease lymphocyte proliferation and promote apoptosis of fibroblasts. At the molecular level, it can decrease expression of HIF, ICAM-1, TNF-α, TGF-β, and IFN-γ, as well as increase vascular VEGF expression and angiogenesis. Thus, we hypothesize that HBO may contribute to treat OPMDs, including oral lichen planus, oral leukoplakia, and oral submucous fibrosis both at the cellular level and the molecular level, and that it would be a safe and inexpensive therapeutic strategy.
Collapse
Affiliation(s)
- Xiaojing Ye
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, PR China
| | - Jing Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, PR China
| | - Rui Lu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, PR China
| | - Gang Zhou
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, PR China; Department of Oral Medicine, School and Hospital of Stomatology, Wuhan University, PR China.
| |
Collapse
|