1
|
Hua Q, Meng X, Chen W, Xu Y, Xu R, Shi Y, Li J, Meng X, Li A, Chai Q, Sheng M, Yao Y, Fan Y, Qiao R, Zhang Y, Wang T, Zhang Y, Cui X, Yu Y, Li H, Tang R, Yan M, Duo B, Dunzhu D, Ga Z, Hou L, Liu Y, Shang J, Chen Q, Qiu X, Ye C, Gong J, Zhu T. Associations of Short-Term Ozone Exposure With Hypoxia and Arterial Stiffness. J Am Coll Cardiol 2025:S0735-1097(24)10559-1. [PMID: 39846938 DOI: 10.1016/j.jacc.2024.11.044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 11/19/2024] [Accepted: 11/21/2024] [Indexed: 01/24/2025]
Abstract
BACKGROUND Epidemiological studies reported associations between ozone (O3) exposure and cardiovascular diseases, yet the biological mechanisms remain underexplored. Hypoxia is a shared pathogenesis of O3-associated diseases; therefore, we hypothesized that O3 exposure may induce changes in hypoxia-related markers, leading to adverse cardiovascular effects. OBJECTIVES This study aimed to investigate associations of short-term O3 exposure with hypoxic biomarkers and arterial stiffness. METHODS We conducted a panel study involving 210 young healthy residents in 2 cities at different altitudes on the Qinghai-Tibetan Plateau in China, where O3 concentrations are high and particulate pollution is low. Participants underwent 4 repeated visits to assess ambient O3 exposure levels, hypoxic biomarkers, and arterial stiffness. We applied linear mixed-effects models to assess the associations of O3 exposure (lag1 to lag1-7 days) with hypoxic biomarkers and arterial stiffness, adjusted for confounders. Mediation analyses explored the hypoxia's role in O3-related arterial stiffness changes. We further examined effect modification by residence altitude and the robustness of results by including PM2.5 (particulate matter ≤2.5 μm in aerodynamic diameter) or NO2 in 2-pollutant models. RESULTS O3 exposure 1 to 7 days before visits was significantly associated with changes in multiple hypoxic biomarkers. A 10-ppb increase in O3 exposure was linked to significant decreases in oxygen saturation (SpO2) and increases in red blood cell count (RBC), hemoglobin concentration, and hematocrit, with maximum changes by -0.42%, 0.92%, 0.97%, and 1.92%, respectively. Laboratory analysis of mRNA and protein markers consistently indicated that O3 exposure activated the hypoxia-inducible factor 1 (HIF-1) signaling pathway. Additionally, a 10-ppb increase in O3 corresponded to a 1.04% to 1.33% increase in carotid-femoral pulse wave velocity (cfPWV), indicating increased arterial stiffness. RBC, hemoglobin concentration, and hematocrit increases significantly mediated the O3-cfPWV association, whereas the SpO2 reduction had an insignificant mediating effect. Associations of O3 with hypoxic biomarkers varied by altitude. The higher altitude group showed delayed associations with SpO₂ and HIF-1 expression but stronger associations with RBC indices. These associations remained robust after adjusting for copollutants. CONCLUSIONS O3 exposure may reduce oxygen availability, prompting compensatory increases in red blood cells and hemoglobin, which exacerbate arterial stiffening. These findings provide new insights into the mechanisms underlying O3-induced cardiovascular injury.
Collapse
Affiliation(s)
- Qiaoyi Hua
- SKL-ESPC & SEPKL-AERM, College of Environmental Sciences and Engineering, Center for Environment and Health, Peking University, Beijing, China
| | - Xin Meng
- SKL-ESPC & SEPKL-AERM, College of Environmental Sciences and Engineering, Center for Environment and Health, Peking University, Beijing, China
| | - Wu Chen
- SKL-ESPC & SEPKL-AERM, College of Environmental Sciences and Engineering, Center for Environment and Health, Peking University, Beijing, China
| | - Yifan Xu
- SKL-ESPC & SEPKL-AERM, College of Environmental Sciences and Engineering, Center for Environment and Health, Peking University, Beijing, China
| | - Ruiwei Xu
- SKL-ESPC & SEPKL-AERM, College of Environmental Sciences and Engineering, Center for Environment and Health, Peking University, Beijing, China
| | - Yunxiu Shi
- SKL-ESPC & SEPKL-AERM, College of Environmental Sciences and Engineering, Center for Environment and Health, Peking University, Beijing, China
| | - Jiajianghui Li
- Institute of Reproductive and Child Health/Ministry of Health Key Laboratory of Reproductive Health and Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China
| | - Xueling Meng
- SKL-ESPC & SEPKL-AERM, College of Environmental Sciences and Engineering, Center for Environment and Health, Peking University, Beijing, China
| | - Ailin Li
- SKL-ESPC & SEPKL-AERM, College of Environmental Sciences and Engineering, Center for Environment and Health, Peking University, Beijing, China
| | - Qianqian Chai
- SKL-ESPC & SEPKL-AERM, College of Environmental Sciences and Engineering, Center for Environment and Health, Peking University, Beijing, China
| | - Mengshuang Sheng
- SKL-ESPC & SEPKL-AERM, College of Environmental Sciences and Engineering, Center for Environment and Health, Peking University, Beijing, China
| | - Yuan Yao
- SKL-ESPC & SEPKL-AERM, College of Environmental Sciences and Engineering, Center for Environment and Health, Peking University, Beijing, China
| | - Yunfei Fan
- SKL-ESPC & SEPKL-AERM, College of Environmental Sciences and Engineering, Center for Environment and Health, Peking University, Beijing, China; China National Environmental Monitoring Centre, Beijing, China
| | - Ruohong Qiao
- SKL-ESPC & SEPKL-AERM, College of Environmental Sciences and Engineering, Center for Environment and Health, Peking University, Beijing, China
| | - Yi Zhang
- SKL-ESPC & SEPKL-AERM, College of Environmental Sciences and Engineering, Center for Environment and Health, Peking University, Beijing, China
| | - Teng Wang
- SKL-ESPC & SEPKL-AERM, College of Environmental Sciences and Engineering, Center for Environment and Health, Peking University, Beijing, China
| | - Yidan Zhang
- SKL-ESPC & SEPKL-AERM, College of Environmental Sciences and Engineering, Center for Environment and Health, Peking University, Beijing, China
| | - Xiaoyu Cui
- SKL-ESPC & SEPKL-AERM, College of Environmental Sciences and Engineering, Center for Environment and Health, Peking University, Beijing, China
| | - Yaqi Yu
- SKL-ESPC & SEPKL-AERM, College of Environmental Sciences and Engineering, Center for Environment and Health, Peking University, Beijing, China
| | - Haonan Li
- SKL-ESPC & SEPKL-AERM, College of Environmental Sciences and Engineering, Center for Environment and Health, Peking University, Beijing, China
| | - Rui Tang
- SKL-ESPC & SEPKL-AERM, College of Environmental Sciences and Engineering, Center for Environment and Health, Peking University, Beijing, China
| | - Meilin Yan
- SKL-ESPC & SEPKL-AERM, College of Environmental Sciences and Engineering, Center for Environment and Health, Peking University, Beijing, China; Department of Environmental Science and Engineering, School of Light Industry Science and Engineering, Beijing Technology and Business University, Beijing, China
| | - Bu Duo
- School of Ecology and Environment, Tibet University, Lhasa, Tibet Autonomous Region, China
| | - Danzeng Dunzhu
- College of Medicine, Tibet University, Lhasa, Tibet Autonomous Region, China; Research Station of Alpine Ecology Environment and Health at Tibet University, Lhasa, Tibet Autonomous Region, China
| | - Zhuo Ga
- Clinical Laboratory, the Second People's Hospital of Tibet Autonomous Region, Lhasa, Tibet Autonomous Region, China
| | - Lei Hou
- College of Resources and Environment, Tibet Agricultural and Animal Husbandry University, Nyingchi, Tibet Autonomous Region, China
| | - Yingjun Liu
- SKL-ESPC & SEPKL-AERM, College of Environmental Sciences and Engineering, Center for Environment and Health, Peking University, Beijing, China; Research Station of Alpine Ecology Environment and Health at Tibet University, Lhasa, Tibet Autonomous Region, China
| | - Jing Shang
- SKL-ESPC & SEPKL-AERM, College of Environmental Sciences and Engineering, Center for Environment and Health, Peking University, Beijing, China; Research Station of Alpine Ecology Environment and Health at Tibet University, Lhasa, Tibet Autonomous Region, China
| | - Qi Chen
- SKL-ESPC & SEPKL-AERM, College of Environmental Sciences and Engineering, Center for Environment and Health, Peking University, Beijing, China
| | - Xinghua Qiu
- SKL-ESPC & SEPKL-AERM, College of Environmental Sciences and Engineering, Center for Environment and Health, Peking University, Beijing, China; Research Station of Alpine Ecology Environment and Health at Tibet University, Lhasa, Tibet Autonomous Region, China
| | - Chunxiang Ye
- SKL-ESPC & SEPKL-AERM, College of Environmental Sciences and Engineering, Center for Environment and Health, Peking University, Beijing, China; Research Station of Alpine Ecology Environment and Health at Tibet University, Lhasa, Tibet Autonomous Region, China
| | - Jicheng Gong
- SKL-ESPC & SEPKL-AERM, College of Environmental Sciences and Engineering, Center for Environment and Health, Peking University, Beijing, China; Research Station of Alpine Ecology Environment and Health at Tibet University, Lhasa, Tibet Autonomous Region, China.
| | - Tong Zhu
- SKL-ESPC & SEPKL-AERM, College of Environmental Sciences and Engineering, Center for Environment and Health, Peking University, Beijing, China; Research Station of Alpine Ecology Environment and Health at Tibet University, Lhasa, Tibet Autonomous Region, China.
| |
Collapse
|
2
|
Ding S, Ding W, Zhang Y, Chen Y, Tang H, Jiang X, Chen J. Serum HIF-1α, IGF-1 and IGFBP-3 correlate to recurrence and overall survival in early-stage hepatocellular carcinoma patients. Biomark Med 2024; 18:1027-1036. [PMID: 39552593 PMCID: PMC11633424 DOI: 10.1080/17520363.2024.2421149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 10/22/2024] [Indexed: 11/19/2024] Open
Abstract
Aim: Recurrence of hepatocellular carcinoma (HCC) after ultrasound-guided microwave ablation (UGMWA) was a critical issue. Therefore, it is significant to identify the role of hypoxia-inducible factor 1 α (HIF-1α), insulin-like growth factor-1 (IGF-1) and IGF binding protein-3 (IGFBP-3) in recurrence.Materials & methods: HCC patients receiving UGMWA were divided into recurrence and no-recurrence groups. The preoperative and postoperative risk factors were compared between these two groups.Results: Preoperative and postoperative serum levels of HIF-1α, IGF-1 and IGFBP-3 were closely associated with the recurrence of HCC. Serum HIF-1α level was increased, while serum levels of IGF-1 and IGFBP-3 were decreased in HCC patients with recurrence.Conclusion: HIF-1α, IGF-1 and IGFBP-3 were associated with and predicted the recurrence of HCC after UGMWA, respectively or in combination.
Collapse
Affiliation(s)
- Shujun Ding
- Department of Ultrasonography, Wuxi People's Hospital Affiliated to Nanjing Medical University, Qingyang Road, Wuxi, 214023, Jiangsu, China
| | - Wei Ding
- Department of Interventional Radiology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Qingyang Road, Wuxi, 214023, Jiangsu, China
| | - Ye Zhang
- Department of General Surgery, Wuxi People's Hospital Affiliated to Nanjing Medical University, Qingyang Road, Wuxi, 214023, Jiangsu, China
| | - Yunbao Chen
- Department of Ultrasonography, Wuxi People's Hospital Affiliated to Nanjing Medical University, Qingyang Road, Wuxi, 214023, Jiangsu, China
| | - Hongtao Tang
- Department of Ultrasonography, Wuxi People's Hospital Affiliated to Nanjing Medical University, Qingyang Road, Wuxi, 214023, Jiangsu, China
| | - Xiao Jiang
- Department of Ultrasonography, Shanghai Yangpu District Shidong Hospital, No. 999 Shiguang Road, Shanghai, 200438, China
| | - Jun Chen
- Department of Ultrasonography, Wuxi People's Hospital Affiliated to Nanjing Medical University, Qingyang Road, Wuxi, 214023, Jiangsu, China
| |
Collapse
|
3
|
Rolver MG, Severin M, Pedersen SF. Regulation of cancer cell lipid metabolism and oxidative phosphorylation by microenvironmental acidosis. Am J Physiol Cell Physiol 2024; 327:C869-C883. [PMID: 39099426 DOI: 10.1152/ajpcell.00429.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 07/30/2024] [Accepted: 07/30/2024] [Indexed: 08/06/2024]
Abstract
The expansion of cancer cell mass in solid tumors generates a harsh environment characterized by dynamically varying levels of acidosis, hypoxia, and nutrient deprivation. Because acidosis inhibits glycolytic metabolism and hypoxia inhibits oxidative phosphorylation, cancer cells that survive and grow in these environments must rewire their metabolism and develop a high degree of metabolic plasticity to meet their energetic and biosynthetic demands. Cancer cells frequently upregulate pathways enabling the uptake and utilization of lipids and other nutrients derived from dead or recruited stromal cells, and in particular lipid uptake is strongly enhanced in acidic microenvironments. The resulting lipid accumulation and increased reliance on β-oxidation and mitochondrial metabolism increase susceptibility to oxidative stress, lipotoxicity, and ferroptosis, in turn driving changes that may mitigate such risks. The spatially and temporally heterogeneous tumor microenvironment thus selects for invasive, metabolically flexible, and resilient cancer cells capable of exploiting their local conditions and of seeking out more favorable surroundings. This phenotype relies on the interplay between metabolism, acidosis, and oncogenic mutations, driving metabolic signaling pathways such as peroxisome proliferator-activated receptors (PPARs). Understanding the particular vulnerabilities of such cells may uncover novel therapeutic liabilities of the most aggressive cancer cells.
Collapse
Affiliation(s)
- Michala G Rolver
- Section for Computational and RNA Biology, Department of Biology, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
- Section for Cell Biology and Physiology, Department of Biology, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Marc Severin
- Section for Computational and RNA Biology, Department of Biology, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
- Section for Cell Biology and Physiology, Department of Biology, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Stine F Pedersen
- Section for Computational and RNA Biology, Department of Biology, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
- Section for Cell Biology and Physiology, Department of Biology, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
4
|
Hazzaa HH, El Shiekh MAM, Elkashty O, Magdy E, Riad D, Khalifa E, Elewa GM, Kamal NM. A critical influence of HIF-1 on MMP-9 and Galectin-3 in oral lichen planus. BMC Oral Health 2024; 24:756. [PMID: 38951854 PMCID: PMC11218350 DOI: 10.1186/s12903-024-04457-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Accepted: 06/06/2024] [Indexed: 07/03/2024] Open
Abstract
OBJECTIVE Oral lichen planus carries a risk for malignancy. The pathogenesis of the disease is mediated by various inflammatory mediators. Several mediators could be responsible for the oncogenic behavior in certain cases. Hypoxia-inducible factor-1a (HIF-1), and its possible correlation to Galactin-3 (Gal-3) and matrix metalloproteinase-9 (MMP-9) over expression represents an important indicator for malignant transformation. The investigation of these factors may present evidence-based information on malignant transformation of the disease. SUBJECTS AND METHODS The study investigated the expression of HIF-1, Gla-3 and MMP-9 in tissue samples of OLP compared to control subjects of un-inflamed gingival overgrowth. 20 biospecimen were allocated in each group. RESULTS Immunohistochemical findings of OLP showed immunoreactivity for Galectin 3, HIF1a and MMP-9 by most of the epithelial cells. There was a positive correlation between HIF1α and MMP-9, r = 0.9301 (P-value < 0.00001). A positive correlation was detected between Galectin 3 and MMP-9, r = 0.7292 (P-value = 0.000264) between Galectin 3 and HIF1α, r = 0.5893 (P-value = 0.006252). CONCLUSION These findings confirm the hypothesis that the adaptive pathways to hypoxia as Gal 3 and MMP-9 expressions and their HIF-1 may play a crucial role in carcinogenesis of OLP.
Collapse
Affiliation(s)
- Hala H Hazzaa
- Department of Oral Medicine, Periodontology and Diagnosis, Faculty of Dental Medicine for Girls, Al Azhar University, Cairo, Egypt
| | - Marwa A M El Shiekh
- Oral and Dental Biology Department, Faculty of Dental Medicine for Girls, Al Azhar University, Cairo, Egypt
| | - Osama Elkashty
- Department of Oral Pathology, Faculty of Dentistry, Mansoura University, Mansoura, Egypt
| | - Eman Magdy
- Department of Oral Medicine, Diagnosis and Periodontology, Faculty of Dentistry, Beni_Suef University, Maadi, Cairo, Egypt.
| | - Dalia Riad
- Department of Oral Biology, Faculty of Dentistry, Beni_Suef University, Maadi, Cairo, Egypt
| | - Eman Khalifa
- Department of Cell Biology, Faculty of Oral and Dental Medicine, Delta University for Science & Technology, Dakhliya, Egypt
| | - Gasser M Elewa
- Department of Oral Medicine, Periodontology, Diagnosis and Oral Radiology, Delta University for Science and Technology, Dakahlia, Egypt
| | - Naglaa M Kamal
- Department of Oral Pathology, Faculty of Oral and Dental Medicine, Ahram Canadian University, 6th of October, Cairo, Egypt
| |
Collapse
|
5
|
Qian L, Zhu Y, Deng C, Liang Z, Chen J, Chen Y, Wang X, Liu Y, Tian Y, Yang Y. Peroxisome proliferator-activated receptor gamma coactivator-1 (PGC-1) family in physiological and pathophysiological process and diseases. Signal Transduct Target Ther 2024; 9:50. [PMID: 38424050 PMCID: PMC10904817 DOI: 10.1038/s41392-024-01756-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 01/13/2024] [Accepted: 01/23/2024] [Indexed: 03/02/2024] Open
Abstract
Peroxisome proliferator-activated receptor gamma coactivator-1 (PGC-1) family (PGC-1s), consisting of three members encompassing PGC-1α, PGC-1β, and PGC-1-related coactivator (PRC), was discovered more than a quarter-century ago. PGC-1s are essential coordinators of many vital cellular events, including mitochondrial functions, oxidative stress, endoplasmic reticulum homeostasis, and inflammation. Accumulating evidence has shown that PGC-1s are implicated in many diseases, such as cancers, cardiac diseases and cardiovascular diseases, neurological disorders, kidney diseases, motor system diseases, and metabolic disorders. Examining the upstream modulators and co-activated partners of PGC-1s and identifying critical biological events modulated by downstream effectors of PGC-1s contribute to the presentation of the elaborate network of PGC-1s. Furthermore, discussing the correlation between PGC-1s and diseases as well as summarizing the therapy targeting PGC-1s helps make individualized and precise intervention methods. In this review, we summarize basic knowledge regarding the PGC-1s family as well as the molecular regulatory network, discuss the physio-pathological roles of PGC-1s in human diseases, review the application of PGC-1s, including the diagnostic and prognostic value of PGC-1s and several therapies in pre-clinical studies, and suggest several directions for future investigations. This review presents the immense potential of targeting PGC-1s in the treatment of diseases and hopefully facilitates the promotion of PGC-1s as new therapeutic targets.
Collapse
Affiliation(s)
- Lu Qian
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Northwest University, Xi'an, 710021, China
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Yanli Zhu
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Northwest University, Xi'an, 710021, China
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Chao Deng
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Zhenxing Liang
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe East, Zhengzhou, 450052, China
| | - Junmin Chen
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Northwest University, Xi'an, 710021, China
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Ying Chen
- Department of Hematology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Xue Wang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, 710061, China
| | - Yanqing Liu
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Northwest University, Xi'an, 710021, China
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Ye Tian
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Northwest University, Xi'an, 710021, China
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710069, China
| | - Yang Yang
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Northwest University, Xi'an, 710021, China.
- Xi'an Key Laboratory of Innovative Drug Research for Heart Failure, Faculty of Life Sciences and Medicine, Northwest University, 229 Taibai North Road, Xi'an, 710069, China.
| |
Collapse
|
6
|
Ray SK, Mukherjee S. Hyperoxic-hypoxic Paradox: Breast Cancer Microenvironment and an Innovative Treatment Strategy. Anticancer Agents Med Chem 2024; 24:729-732. [PMID: 38415470 DOI: 10.2174/0118715206290816240220062545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 02/02/2024] [Accepted: 02/06/2024] [Indexed: 02/29/2024]
Abstract
A small therapeutic range of oxygen is required for effective metabolism. As a result, hypoxia (low oxygen concentration) is one of the most potent inducers of gene expression, metabolic alterations, and regenerative processes, such as angiogenesis, stem cell proliferation, migration, and differentiation. The cellular response is controlled by sensing the increased oxygen levels (hyperoxia) or hypoxia via specific chemoreceptor cells. Surprisingly, changes in free oxygen concentration instead of absolute oxygen levels may be regarded as a deficiency of oxygen at the cellular level. Recurrent intermittent hyperoxia may trigger many mediators of cellular pathways typically generated during hypoxia. The dilemma of hyperoxic-hypoxic conditions is known as the hyperoxic-hypoxic paradox. According to the latest data, the hypoxic microenvironment, crucial during cancer formation, has been demonstrated to play a key role in regulating breast cancer growth and metastasis. Hypoxic circumstances cause breast cancer cells to respond in a variety of ways. Transcription factors are identified as hypoxia-inducible factors (HIFs) that have been suggested to be a factor in the pathobiology of breast cancer and a possible therapeutic target, driving the cellular response to hypoxia. Breast cancer has a dismal prognosis due to a high level of resistance to practically all well-known cancer management that has been related to hypoxia-based interactions between tumor cells and the stromal milieu. We attempt to review the enigma by exploring the starring roles of HIFs in breast cancer, the HIF paradox, and the hyperoxic-hypoxic enigma.
Collapse
Affiliation(s)
- Suman Kumar Ray
- Independent Researcher, Bhopal, Madhya Pradesh, 462020, India
| | - Sukhes Mukherjee
- Department of Biochemistry, All India Institute of Medical Science, Bhopal, Madhya Pradesh, 462020, India
| |
Collapse
|
7
|
Tian Y, Zhao L, Gui Z, Liu S, Liu C, Yu T, Zhang L. PI3K/AKT signaling activates HIF1α to modulate the biological effects of invasive breast cancer with microcalcification. NPJ Breast Cancer 2023; 9:93. [PMID: 37957150 PMCID: PMC10643473 DOI: 10.1038/s41523-023-00598-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 10/20/2023] [Indexed: 11/15/2023] Open
Abstract
Microcalcification (MC) is a valuable diagnostic indicator of breast cancer, and it is reported to be associated with increased tumor aggressiveness and poor prognosis. Nevertheless, the exact potential molecular mechanism is not completely understood. Here, we find that the mineralized invasive breast cancer (IBC) cells not only increased their proliferation and migration, but also showed the characteristic of doxorubicin resistance. The PI3K/AKT signaling pathway is associated with the generation of calcification in IBC, and it activates the transcription and translation of its downstream hypoxia-inducible factor 1α (HIF1α). Knockdown of HIF1α protein significantly downregulated cell proliferation and migration while calcification persists. Meanwhile, calcified breast cancer cells restored sensitivity to doxorubicin because of suppressed HIF1α expression. In addition, we provide initial data on the underlying value of HIF1α as a biomarker of doxorubicin resistance. These findings provide a new direction for exploring microcalcifications in IBC.
Collapse
Affiliation(s)
- Yao Tian
- Department of Thyroid and Breast Surgery, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei Province, 430030, China
| | - Lu Zhao
- Department of Thyroid and Breast Surgery, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei Province, 430030, China
| | - Zhengwei Gui
- Department of Thyroid and Breast Surgery, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei Province, 430030, China
| | - Shiyang Liu
- Department of Thyroid and Breast Surgery, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei Province, 430030, China
| | - Chenguang Liu
- Department of Thyroid and Breast Surgery, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei Province, 430030, China
| | - Tianyao Yu
- Department of Thyroid and Breast Surgery, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei Province, 430030, China
| | - Lin Zhang
- Department of Thyroid and Breast Surgery, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei Province, 430030, China.
| |
Collapse
|
8
|
Ganesan K, Xu C, Liu Q, Sui Y, Chen J. Unraveling the Role of Hepatic PGC1α in Breast Cancer Invasion: A New Target for Therapeutic Intervention? Cells 2023; 12:2311. [PMID: 37759533 PMCID: PMC10529029 DOI: 10.3390/cells12182311] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 09/15/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023] Open
Abstract
Breast cancer (BC) is the most common cancer among women worldwide and the main cause of cancer deaths in women. Metabolic components are key risk factors for the development of non-alcoholic fatty liver disease (NAFLD), which may promote BC. Studies have reported that increasing PGC1α levels increases mitochondrial biogenesis, thereby increasing cell proliferation and metastasis. Moreover, the PGC1α/ERRα axis is a crucial regulator of cellular metabolism in various tissues, including BC. However, it remains unclear whether NAFLD is closely associated with the risk of BC. Therefore, the present study aimed to determine whether hepatic PGC1α promotes BC cell invasion via ERRα. Various assays, including ELISA, western blotting, and immunoprecipitation, have been employed to explore these mechanisms. According to the KM plot and TCGA data, elevated PGC1α expression was highly associated with a shorter overall survival time in patients with BC. High concentrations of palmitic acid (PA) promoted PGC1α expression, lipogenesis, and inflammatory processes in hepatocytes. Conditioned medium obtained from PA-treated hepatocytes significantly increased BC cell proliferation. Similarly, recombinant PGC1α in E0771 and MCF7 cells promoted cell proliferation, migration, and invasion in vitro. However, silencing PGC1α in both BC cell lines resulted in a decrease in this trend. As determined by immunoprecipitation assay, PCG1a interacted with ERRα, thereby facilitating the proliferation of BC cells. This outcome recognizes the importance of further investigations in exploring the full potential of hepatic PGC1α as a prognostic marker for BC development.
Collapse
Affiliation(s)
- Kumar Ganesan
- School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Sassoon Road, Hong Kong, China; (K.G.); (C.X.); (Q.L.); (Y.S.)
| | - Cong Xu
- School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Sassoon Road, Hong Kong, China; (K.G.); (C.X.); (Q.L.); (Y.S.)
| | - Qingqing Liu
- School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Sassoon Road, Hong Kong, China; (K.G.); (C.X.); (Q.L.); (Y.S.)
| | - Yue Sui
- School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Sassoon Road, Hong Kong, China; (K.G.); (C.X.); (Q.L.); (Y.S.)
| | - Jianping Chen
- School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Sassoon Road, Hong Kong, China; (K.G.); (C.X.); (Q.L.); (Y.S.)
- Shenzhen Institute of Research and Innovation, The University of Hong Kong, Shenzhen 518057, China
| |
Collapse
|
9
|
Mirzaei S, Ranjbar B, Tackallou SH, Aref AR. Hypoxia inducible factor-1α (HIF-1α) in breast cancer: The crosstalk with oncogenic and onco-suppressor factors in regulation of cancer hallmarks. Pathol Res Pract 2023; 248:154676. [PMID: 37454494 DOI: 10.1016/j.prp.2023.154676] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 07/03/2023] [Accepted: 07/04/2023] [Indexed: 07/18/2023]
Abstract
Low oxygen level at tumor microenvironment leads to a condition, known as hypoxia that is implicated in cancer progression. Upon hypoxia, HIF-1α undergoes activation and due to its oncogenic function and interaction with other molecular pathways, promotes tumor progression. The HIF-1α role in regulating breast cancer progression is described, Overall, HIF-1α has upregulation in breast tumor and due to its tumor-promoting function, its upregulation is in favor of breast tumor progression. HIF-1α overexpression prevents apoptosis in breast tumor and it promotes cell cycle progression. Silencing HIF-1α triggers cycle arrest and decreases growth. Migration of breast tumor enhances by HIF-1α signaling and it mainly induces EMT in providing metastasis. HIF-1α upregulation stimulates drug resistance and radio-resistance in breast tumor. Furthermore, HIF-1α signaling induces immune evasion of breast cancer. Berberine and pharmacological intervention suppress HIF-1α signaling in breast tumor and regulation of HIF-1α by non-coding RNAs occurs. Furthermore, HIF-1α is a biomarker in clinic.
Collapse
Affiliation(s)
- Sepideh Mirzaei
- Department of Biology, Faculty of Science, Islamic Azad University, Science and Research Branch, Tehran, Iran.
| | - Bijan Ranjbar
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran 14117-13116, Iran
| | | | - Amir Reza Aref
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02115, USA
| |
Collapse
|
10
|
Calahorra J, Araujo-Abad S, Granadino-Roldán JM, Naranjo Á, Martínez-Lara E, Siles E. Tyrosol: Repercussion of the Lack of a Hydroxyl-Group in the Response of MCF-7 Cells to Hypoxia. J Med Food 2023. [PMID: 37379464 DOI: 10.1089/jmf.2022.0096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/30/2023] Open
Abstract
In solid tumors, such as breast cancer, hypoxic microenvironment worsens patient prognoses. We have previously reported in MCF-7 breast cancer cells that, under hypoxic conditions, hydroxytyrosol (HT) downregulates the level of reactive oxygen species, reduces the expression of hypoxia inducible factor-1α (HIF-1α), and, at high concentrations, can bind to the aryl hydrocarbon receptor (AhR). With this background, the present study investigated whether the most abundant extra virgin olive oil (EVOO) phenolic compound tyrosol (TYR), with a chemical structure similar to HT but with only one hydroxyl group, exerts comparable effects. Our results revealed that, although TYR did not show any antioxidant activity in hypoxic MCF-7 cells, it inhibited the PI3K/Akt/mTOR/S6 kinase (S6K) pathway and reduced the expression of HIF-1α and some of its target genes. Besides, TYR showed a lower binding affinity with the cytosolic transcription factor AhR, and even reduced its transcriptional activity. Some of these results are positive to control tumor progression in a hypoxic environment; however, they are observed at doses unachievable with diet intake or nutraceutical presentations. Considering that EVOO phenols can have synergistic effects, a mixture of low doses of TYR and other phenols could be useful to achieve these beneficial effects.
Collapse
Affiliation(s)
- Jesús Calahorra
- Departamento de Biología Experimental, Universidad de Jaén, Jaén, Spain
| | | | | | - Ángela Naranjo
- Departamento de Biología Experimental, Universidad de Jaén, Jaén, Spain
| | | | - Eva Siles
- Departamento de Biología Experimental, Universidad de Jaén, Jaén, Spain
| |
Collapse
|
11
|
Jin Y, Bian S, Wang H, Mo J, Fei H, Li L, Chen T, Jiang H. CRMP2 derived from cancer associated fibroblasts facilitates progression of ovarian cancer via HIF-1α-glycolysis signaling pathway. Cell Death Dis 2022; 13:675. [PMID: 35927239 PMCID: PMC9352901 DOI: 10.1038/s41419-022-05129-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 07/18/2022] [Accepted: 07/22/2022] [Indexed: 01/21/2023]
Abstract
As the predominant stroma cells of tumor microenvironment (TME), cancer associated fibroblasts (CAFs) are robust tumor player of different malignancies. However, less is known about the regulatory mechanism of CAFs on promoting progression of ovarian cancer (OvCA). In the present study, the conditioned medium of primary CAFs (CAF-CM) from OvCA was used to culture cell lines of epithelial ovarian cancer (EOC), and showed a potent role in promoting proliferation, migration and invasion of cancer cells. Mass spectrum (MS) analysis identified that Collapsin response mediator protein-2 (CRMP2), a microtubule-associated protein involved in diverse malignancies, derived from CAFs was a key regulator responsible for mediating these cell events of OvCA. In vitro study using recombinant CRMP2 (r-CRMP2) revealed that the protein promoted proliferation, invasion, and migration of OvCA cells through activation of hypoxia-inducible factor (HIF)-1α-glycolysis signaling pathway. The CRMP2 was abundantly expressed in OvCA, with a well correlation with metastasis and poor prognosis, as analyzed from 118 patients' samples. Inhibition of the CRMP2 derived from CAFs by neutralizing antibodies significantly attenuated the tumor size, weights, and metastatic foci numbers of mice in vivo. Our finding has provided a novel therapeutic clue for OvCA based on TME.
Collapse
Affiliation(s)
- Yunfeng Jin
- grid.412312.70000 0004 1755 1415Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011 China ,grid.440642.00000 0004 0644 5481Department of Obstetrics and Gynecology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001 China
| | - Saiyan Bian
- grid.440642.00000 0004 0644 5481Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001 China
| | - Hui Wang
- grid.412312.70000 0004 1755 1415Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011 China
| | - Jiahang Mo
- grid.412312.70000 0004 1755 1415Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011 China
| | - He Fei
- grid.412312.70000 0004 1755 1415Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011 China
| | - Li Li
- grid.440642.00000 0004 0644 5481Department of Obstetrics and Gynecology, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001 China
| | - Tong Chen
- grid.8547.e0000 0001 0125 2443Department of Hematology, Huashan Hospital, Fudan University, Shanghai, 200040 China
| | - Hua Jiang
- grid.412312.70000 0004 1755 1415Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011 China
| |
Collapse
|
12
|
Ansari MI, Bano N, Kainat KM, Singh VK, Sharma PK. Bisphenol A exposure induces metastatic aggression in low metastatic MCF-7 cells via PGC-1α mediated mitochondrial biogenesis and epithelial-mesenchymal plasticity. Life Sci 2022; 302:120649. [PMID: 35597549 DOI: 10.1016/j.lfs.2022.120649] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 05/06/2022] [Accepted: 05/14/2022] [Indexed: 11/26/2022]
Abstract
AIMS The frequency of estrogen receptor alpha (ERα)-positive breast cancers and their metastatic progression is prevalent in females globally. Aberrant interaction of estrogen-like endocrine-disrupting chemicals (EDCs) is highly implicated in breast carcinogenesis. Studies have shown that single or acute exposures of weak EDCs such as bisphenol A (BPA) may not have a substantial pro-carcinogenic/metastatic effect. However, repeated exposure to EDCs is expected to strongly induce carcinogenic/metastatic progression, which remains to be studied. MAIN METHODS Low metastatic ERα-positive human breast cancer cells (MCF-7) were exposed to nanomolar doses of BPA every 24 h (up to 200 days) to study the effect of repeated exposure on metastatic potential. Following the designated treatment of BPA, markers of epithelial-mesenchymal transition (EMT), migration and invasion, mitochondrial biogenesis, ATP levels, and peroxisome proliferator-activated receptor-gamma coactivator 1-alpha (PGC-1α) knockdown assays were performed. KEY FINDINGS A repeated exposure of low dose BPA induced stable epithelial-mesenchymal plasticity in MCF-7 cells to augment migration and invasion in the ERα-dependent pathway. Repeated exposures of BPA increased the levels of several mesenchymal markers such as N-cadherin, vimentin, cluster of differentiation 44 (CD44), slug, and alpha-smooth muscle actin (α-SMA), whereas reduced the level of E-cadherin drastically. BPA-induced mitochondrial biogenesis favored metastatic aggression by fulfilling bioenergetics demand via PGC-1α/NRF1/ERRα signaling. Knockdown of PGC-1α resulted in suppressing both mitochondrial biogenesis and EMT in BPA exposed MCF-7 cells. SIGNIFICANCE Repeated exposures of low dose BPA may induce metastatic aggression in ERα-positive breast cancer cells via PGC-1α-mediated mitochondrial biogenesis and epithelial-mesenchymal plasticity.
Collapse
Affiliation(s)
- Mohammad Imran Ansari
- Food Drug and Chemical Toxicology Group, CSIR-Indian Institute of Toxicology Research, Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, Uttar Pradesh 226001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Nuzhat Bano
- Food Drug and Chemical Toxicology Group, CSIR-Indian Institute of Toxicology Research, Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, Uttar Pradesh 226001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - K M Kainat
- Food Drug and Chemical Toxicology Group, CSIR-Indian Institute of Toxicology Research, Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, Uttar Pradesh 226001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Vipendra Kumar Singh
- Food Drug and Chemical Toxicology Group, CSIR-Indian Institute of Toxicology Research, Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, Uttar Pradesh 226001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Pradeep Kumar Sharma
- Food Drug and Chemical Toxicology Group, CSIR-Indian Institute of Toxicology Research, Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, Uttar Pradesh 226001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
13
|
miR-138-5p Inhibits Vascular Mimicry by Targeting the HIF-1α/VEGFA Pathway in Hepatocellular Carcinoma. J Immunol Res 2022; 2022:7318950. [PMID: 35669101 PMCID: PMC9167126 DOI: 10.1155/2022/7318950] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 04/12/2022] [Accepted: 04/18/2022] [Indexed: 12/24/2022] Open
Abstract
Tumour vascular mimicry (VM) is the process by which new blood vessels are formed by tumour cells rather than endothelial cells. An increasing number of studies have revealed that the VM process is associated with cancer progression and metastasis. MiR-138-5p has been reported to act as a tumour suppressor in many cancers. However, the role and underlying mechanism of miR-138-5p in hepatocellular carcinoma (HCC) VM remain unclear. In this study, VM density was detected by CD31/periodic acid-Schiff double staining in HCC clinical specimens. We found that miR-138-5p expression correlated strongly and negatively with microvessel density. Additionally, the miR-138-5p mimic or inhibitor decreased or increased, respectively, tube formation capacity in HepG2 and Hep3B cells. Consistent with this finding, miR-138-5p repressed vessel density in vivo. Moreover, miR-138-5p targeted hypoxia-inducible factor 1α (HIF-1α) and regulated the expression of HIF-1α and vascular endothelial growth factor A (VEGFA), which are established classical master regulators for angiogenesis. Consistent with these findings, the HIF-1α inhibitor CAY10585 effectively blocked HCC cell VM and VEGFA expression. In conclusion, miR-138-5p inhibits HepG2 and Hep3B cell VM by blocking the HIF-1α/VEGFA pathway. Therefore, miR-138-5p may serve as a useful therapeutic target for miRNA-based HCC therapy.
Collapse
|
14
|
Urra FA, Fuentes-Retamal S, Palominos C, Rodríguez-Lucart YA, López-Torres C, Araya-Maturana R. Extracellular Matrix Signals as Drivers of Mitochondrial Bioenergetics and Metabolic Plasticity of Cancer Cells During Metastasis. Front Cell Dev Biol 2021; 9:751301. [PMID: 34733852 PMCID: PMC8558415 DOI: 10.3389/fcell.2021.751301] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 09/28/2021] [Indexed: 12/16/2022] Open
Abstract
The role of metabolism in tumor growth and chemoresistance has received considerable attention, however, the contribution of mitochondrial bioenergetics in migration, invasion, and metastasis is recently being understood. Migrating cancer cells adapt their energy needs to fluctuating changes in the microenvironment, exhibiting high metabolic plasticity. This occurs due to dynamic changes in the contributions of metabolic pathways to promote localized ATP production in lamellipodia and control signaling mediated by mitochondrial reactive oxygen species. Recent evidence has shown that metabolic shifts toward a mitochondrial metabolism based on the reductive carboxylation, glutaminolysis, and phosphocreatine-creatine kinase pathways promote resistance to anoikis, migration, and invasion in cancer cells. The PGC1a-driven metabolic adaptations with increased electron transport chain activity and superoxide levels are essential for metastasis in several cancer models. Notably, these metabolic changes can be determined by the composition and density of the extracellular matrix (ECM). ECM stiffness, integrins, and small Rho GTPases promote mitochondrial fragmentation, mitochondrial localization in focal adhesion complexes, and metabolic plasticity, supporting enhanced migration and metastasis. Here, we discuss the role of ECM in regulating mitochondrial metabolism during migration and metastasis, highlighting the therapeutic potential of compounds affecting mitochondrial function and selectively block cancer cell migration.
Collapse
Affiliation(s)
- Félix A Urra
- Laboratorio de Plasticidad Metabólica y Bioenergética, Programa de Farmacología Molecular y Clínica, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile.,Network for Snake Venom Research and Drug Discovery, Santiago, Chile
| | - Sebastián Fuentes-Retamal
- Laboratorio de Plasticidad Metabólica y Bioenergética, Programa de Farmacología Molecular y Clínica, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile.,Network for Snake Venom Research and Drug Discovery, Santiago, Chile
| | - Charlotte Palominos
- Laboratorio de Plasticidad Metabólica y Bioenergética, Programa de Farmacología Molecular y Clínica, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile.,Network for Snake Venom Research and Drug Discovery, Santiago, Chile
| | - Yarcely A Rodríguez-Lucart
- Network for Snake Venom Research and Drug Discovery, Santiago, Chile.,Instituto de Química de Recursos Naturales, Universidad de Talca, Talca, Chile
| | - Camila López-Torres
- Laboratorio de Plasticidad Metabólica y Bioenergética, Programa de Farmacología Molecular y Clínica, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile.,Network for Snake Venom Research and Drug Discovery, Santiago, Chile
| | - Ramiro Araya-Maturana
- Network for Snake Venom Research and Drug Discovery, Santiago, Chile.,Instituto de Química de Recursos Naturales, Universidad de Talca, Talca, Chile
| |
Collapse
|
15
|
Zhou HC, Xin-Yan Yan, Yu WW, Liang XQ, Du XY, Liu ZC, Long JP, Zhao GH, Liu HB. Lactic acid in macrophage polarization: The significant role in inflammation and cancer. Int Rev Immunol 2021; 41:4-18. [PMID: 34304685 DOI: 10.1080/08830185.2021.1955876] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Metabolite lactic acid has always been regarded as a metabolic by-product rather than a bioactive molecule. Recently, this view has changed since it was discovered that lactic acid can be used as a signal molecule and has novel signal transduction functions both intracellular and extracellular, which can regulate key functions in the immune system. In recent years, more and more evidence has shown that lactic acid is closely related to the metabolism and polarization of macrophages. During inflammation, lactic acid is a regulator of macrophage metabolism, and it can prevent excessive inflammatory responses; In malignant tumors, lactic acid produced by tumor tissues promotes the polarization of tumor-associated macrophages, which in turn promotes tumor progression. In this review, we examined the relationship between lactic acid and macrophage metabolism. We further discussed how lactic acid plays a role in maintaining the homeostasis of macrophages, as well as the biology of macrophage polarization and the M1/M2 imbalance in human diseases. Potential methods to target lactic acid in the treatment of inflammation and cancer will also be discussed so as to provide new strategies for the treatment of diseases.
Collapse
Affiliation(s)
- Hai-Cun Zhou
- Department of Breast Surgery, Gansu Maternal and Child Health Care Hospital, Lanzhou, Gansu Province, P. R. China.,Key Laboratory of Stem Cells and Gene Drugs of Gansu Province, The 940th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou, P.R. China.,The Second Clinical Medical College, Lanzhou University, Lanzhou, P.R.China
| | - Xin-Yan Yan
- Department of Breast Surgery, Gansu Maternal and Child Health Care Hospital, Lanzhou, Gansu Province, P. R. China
| | - Wen-Wen Yu
- Key Laboratory of Stem Cells and Gene Drugs of Gansu Province, The 940th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou, P.R. China.,The Second Clinical Medical College, Lanzhou University, Lanzhou, P.R.China
| | - Xiao-Qin Liang
- Key Laboratory of Stem Cells and Gene Drugs of Gansu Province, The 940th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou, P.R. China.,The Second Clinical Medical College, Lanzhou University, Lanzhou, P.R.China
| | - Xiao-Yan Du
- Department of Breast Surgery, Gansu Maternal and Child Health Care Hospital, Lanzhou, Gansu Province, P. R. China
| | - Zhi-Chang Liu
- Key Laboratory of Stem Cells and Gene Drugs of Gansu Province, The 940th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou, P.R. China.,The Second Clinical Medical College, Lanzhou University, Lanzhou, P.R.China
| | - Jian-Ping Long
- Department of Breast Surgery, Gansu Maternal and Child Health Care Hospital, Lanzhou, Gansu Province, P. R. China
| | - Guang-Hui Zhao
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, P. R. China
| | - Hong-Bin Liu
- Key Laboratory of Stem Cells and Gene Drugs of Gansu Province, The 940th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou, P.R. China.,The Second Clinical Medical College, Lanzhou University, Lanzhou, P.R.China
| |
Collapse
|
16
|
Vallespi MG, Mestre B, Marrero MA, Uranga R, Rey D, Lugiollo M, Betancourt M, Silva K, Corrales D, Lamadrid Y, Rodriguez Y, Maceo A, Chaviano PP, Lemos G, Cabrales A, Freyre FM, Santana H, Garay HE, Oliva B, Fernandez JR. A first-in-class, first-in-human, phase I trial of CIGB-552, a synthetic peptide targeting COMMD1 to inhibit the oncogenic activity of NF-κB in patients with advanced solid tumors. Int J Cancer 2021; 149:1313-1321. [PMID: 34019700 DOI: 10.1002/ijc.33695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 04/05/2021] [Accepted: 04/29/2021] [Indexed: 11/06/2022]
Abstract
CIGB-552 is a synthetic peptide that interacts with COMMD1 and upregulates its protein levels. The objectives of this phase I study were safety, pharmacokinetic profile, evaluation of the lymphocytes CD4+ and CD8+ and preliminary activity in patients with advanced tumors. A 3 + 3 dose-escalation design with seven dose levels was implemented. Patients were included until a grade 3 related adverse event occurred and the maximum tolerated dose was reached. The patients received subcutaneous administration of CIGB-552 three times per week for 2 weeks. Single-dose plasma pharmacokinetics was characterized at two dose levels, and tumor responses were classified by RECIST 1.1. Twenty-four patients received CIGB-552. Dose-limiting toxicity was associated with a transient grade 3 pruritic maculopapular rash at a dose of 7.0 mg. The maximum tolerated dose was defined as 4.7 mg. Ten patients were assessable for immunological status. Seven patients had significant changes in the ratio CD4/CD8 in response to CIGB-552 treatment; three patients did not modify the immunological status. Stable disease was observed in five patients, including two metastatic soft sarcomas. We conclude that CIGB-552 at dose 4.7 mg was well tolerated with no significant adverse events and appeared to provide some clinical benefits.
Collapse
Affiliation(s)
| | - Braulio Mestre
- National Institute of Oncology and Radiobiology (INOR), Havana, Cuba
| | - Maria A Marrero
- National Coordinating Center for Clinical Trials (CENCEC), Havana, Cuba
| | - Rolando Uranga
- National Coordinating Center for Clinical Trials (CENCEC), Havana, Cuba
| | - Diana Rey
- National Coordinating Center for Clinical Trials (CENCEC), Havana, Cuba
| | - Martha Lugiollo
- National Institute of Oncology and Radiobiology (INOR), Havana, Cuba
| | - Mircea Betancourt
- National Institute of Oncology and Radiobiology (INOR), Havana, Cuba
| | - Kirenia Silva
- National Institute of Oncology and Radiobiology (INOR), Havana, Cuba
| | - Danay Corrales
- National Institute of Oncology and Radiobiology (INOR), Havana, Cuba
| | - Yanet Lamadrid
- National Institute of Oncology and Radiobiology (INOR), Havana, Cuba
| | - Yamilka Rodriguez
- National Institute of Oncology and Radiobiology (INOR), Havana, Cuba
| | - Anaelys Maceo
- National Coordinating Center for Clinical Trials (CENCEC), Havana, Cuba
| | - Pedro P Chaviano
- National Coordinating Center for Clinical Trials (CENCEC), Havana, Cuba
| | - Gilda Lemos
- Center for Genetic Engineering and Biotechnology (CIGB), Havana, Cuba
| | - Ania Cabrales
- Center for Genetic Engineering and Biotechnology (CIGB), Havana, Cuba
| | - Freya M Freyre
- Center for Genetic Engineering and Biotechnology (CIGB), Havana, Cuba
| | - Hector Santana
- Center for Genetic Engineering and Biotechnology (CIGB), Havana, Cuba
| | - Hilda E Garay
- Center for Genetic Engineering and Biotechnology (CIGB), Havana, Cuba
| | - Brizaida Oliva
- Center for Genetic Engineering and Biotechnology (CIGB), Havana, Cuba
| | - Julio R Fernandez
- Center for Genetic Engineering and Biotechnology (CIGB), Havana, Cuba
| |
Collapse
|
17
|
Špaková I, Rabajdová M, Mičková H, Graier WF, Mareková M. Effect of hypoxia factors gene silencing on ROS production and metabolic status of A375 malignant melanoma cells. Sci Rep 2021; 11:10325. [PMID: 33990669 PMCID: PMC8121821 DOI: 10.1038/s41598-021-89792-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 04/29/2021] [Indexed: 01/04/2023] Open
Abstract
The innate response of melanocytes to exogenous or endogenous stress stimuli like extreme pH and temperature, metabolite and oxygen deficiency or a high UV dose initiates a cellular stress response. This process activates adaptive processes to minimize the negative impact of the stressor on the pigment cell. Under physiological conditions, a non-cancer cell is directed to apoptosis if the stressor persists. However, malignant melanoma cells will survive persistent stress thanks to distinct "cancerous" signaling pathways (e.g. MEK) and transcription factors that regulate the expression of so-called "survival genes" (e.g. HIF, MITF). In this survival response of cancer cells, MEK pathway directs melanoma cells to deregulate mitochondrial metabolism, to accumulate reduced species (NADH), and to centralize metabolism in the cytosol. The aim of this work was to study the effect of gene silencing in malignant melanoma A375 cells on metabolic processes in cytosol and mitochondria. Gene silencing of HIF-1α, and miR-210 in normoxia and pseudohypoxia, and analysis of its effect on MITF-M, and PDHA1 expression. Detection of cytosolic NADH by Peredox-mCherry Assay. Detection of OCR, and ECAR using Seahorse XF96. Measurement of produced O2•- with MitoTracker Red CMXRos. 1H NMR analysis of metabolites present in cell suspension, and medium. By gene silencing of HIF-1α and miR-210 the expression of PDHA1 was upregulated while that of MITF-M was downregulated, yielding acceleration of mitochondrial respiratory activity and thus elimination of ROS. Hence, we detected a significantly reduced A375 cell viability, an increase in alanine, inositol, nucleotides, and other metabolites that together define apoptosis. Based on the results of measurements of mitochondrial resipiratory activity, ROS production, and changes in the metabolites obtained in cells under the observed conditions, we concluded that silencing of HIF-1α and miR-210 yields apoptosis and, ultimately, apoptotic cell death in A375 melanoma cells.
Collapse
Affiliation(s)
- Ivana Špaková
- Department of Medical and Clinical Biochemistry, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, Trieda SNP 1, 04011, Košice, Slovakia
| | - Miroslava Rabajdová
- Department of Medical and Clinical Biochemistry, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, Trieda SNP 1, 04011, Košice, Slovakia.
| | - Helena Mičková
- Department of Biology, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, Košice, Slovakia
| | - Wolfgang F Graier
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria.,BioTechMed, Graz, Austria
| | - Mária Mareková
- Department of Medical and Clinical Biochemistry, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, Trieda SNP 1, 04011, Košice, Slovakia
| |
Collapse
|
18
|
Zhang Y, Zhang H, Wang M, Schmid T, Xin Z, Kozhuharova L, Yu WK, Huang Y, Cai F, Biskup E. Hypoxia in Breast Cancer-Scientific Translation to Therapeutic and Diagnostic Clinical Applications. Front Oncol 2021; 11:652266. [PMID: 33777815 PMCID: PMC7991906 DOI: 10.3389/fonc.2021.652266] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 02/12/2021] [Indexed: 12/13/2022] Open
Abstract
Breast cancer has been the leading cause of female cancer deaths for decades. Intratumoral hypoxia, mainly caused by structural and functional abnormalities in microvasculature, is often associated with a more aggressive phenotype, increased risk of metastasis and resistance to anti-malignancy treatments. The response of cancer cells to hypoxia is ascribed to hypoxia-inducible factors (HIFs) that activate the transcription of a large battery of genes encoding proteins promoting primary tumor vascularization and growth, stromal cell recruitment, extracellular matrix remodeling, cell motility, local tissue invasion, metastasis, and maintenance of the cancer stem cell properties. In this review, we summarized the role of hypoxia specifically in breast cancer, discuss the prognostic and predictive value of hypoxia factors, potential links of hypoxia and endocrine resistance, cancer hypoxia measurements, further involved mechanisms, clinical application of hypoxia-related treatments and open questions.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hongyi Zhang
- Department of Breast Surgery, Yangpu Hospital, Tongji University School of Medicine, Shanghai, China
| | - Minghong Wang
- Department of Health Management, Shanghai Public Health Clinical Center, Shanghai, China
| | - Thomas Schmid
- Department of Medical Oncology, St. Claraspital, Basel, Switzerland
| | - Zhaochen Xin
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | | | - Wai-Kin Yu
- Cellomics International Limited, Hong Kong, China
| | - Yuan Huang
- Cellomics International Limited, Hong Kong, China
| | - Fengfeng Cai
- Department of Breast Surgery, Yangpu Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ewelina Biskup
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Division of Internal Medicine, University Hospital of Basel, University of Basel, Basel, Switzerland.,Department of Advanced Biomedical Sciences, Federico II University of Naples, Naples, Italy
| |
Collapse
|
19
|
Shamis SAK, McMillan DC, Edwards J. The relationship between hypoxia-inducible factor 1α (HIF-1α) and patient survival in breast cancer: Systematic review and meta-analysis. Crit Rev Oncol Hematol 2021; 159:103231. [PMID: 33482350 DOI: 10.1016/j.critrevonc.2021.103231] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 01/06/2021] [Accepted: 01/16/2021] [Indexed: 01/16/2023] Open
Abstract
INTRODUCTION Hypoxia is a characteristic of many solid tumours and results in an increase in expression of HIF-1α. Many studies have investigated the prognostic value of HIF-1α expression in breast cancer (BC), however, the prognostic value remains unclear. Therefore, a systematic review and meta-analysis was undertaken to determine the prognostic value of HIF-1α in BC patients. METHODS The electronic databases PubMed and Web of science were systematically searched to identify relevant papers. The clinical outcomes included disease-free survival (DFS), recurrence-free survival (RFS) and overall survival (OS) in BC patients. Review Manager version 5.4 was employed to analysis data from 30 eligible studies (containing 6201patients). RESULTS High expression of HIF-1α was associated with poorer DFS and OS. There was an effect of survival analysis, study region, antibodies used, scoring and threshold methods on HIF-1α expression. CONCLUSION HIF-1α overexpression was significantly associated with poorer DFS and OS in breast cancer patients.
Collapse
Affiliation(s)
- Suad A K Shamis
- Academic Unit of Surgery, College of Medical, Veterinary and Life Sciences- University of Glasgow, Royal Infirmary, Glasgow, UK; Unit of Experimental Therapeutics, Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences-University of Glasgow, Glasgow, UK.
| | - Donald C McMillan
- Academic Unit of Surgery, College of Medical, Veterinary and Life Sciences- University of Glasgow, Royal Infirmary, Glasgow, UK
| | - Joanne Edwards
- Unit of Experimental Therapeutics, Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences-University of Glasgow, Glasgow, UK
| |
Collapse
|
20
|
Cancer Stem Cell-Associated Pathways in the Metabolic Reprogramming of Breast Cancer. Int J Mol Sci 2020; 21:ijms21239125. [PMID: 33266219 PMCID: PMC7730588 DOI: 10.3390/ijms21239125] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 11/25/2020] [Accepted: 11/26/2020] [Indexed: 02/07/2023] Open
Abstract
Metabolic reprogramming of cancer is now considered a hallmark of many malignant tumors, including breast cancer, which remains the most commonly diagnosed cancer in women all over the world. One of the main challenges for the effective treatment of breast cancer emanates from the existence of a subpopulation of tumor-initiating cells, known as cancer stem cells (CSCs). Over the years, several pathways involved in the regulation of CSCs have been identified and characterized. Recent research has also shown that CSCs are capable of adopting a metabolic flexibility to survive under various stressors, contributing to chemo-resistance, metastasis, and disease relapse. This review summarizes the links between the metabolic adaptations of breast cancer cells and CSC-associated pathways. Identification of the drivers capable of the metabolic rewiring in breast cancer cells and CSCs and the signaling pathways contributing to metabolic flexibility may lead to the development of effective therapeutic strategies. This review also covers the role of these metabolic adaptation in conferring drug resistance and metastasis in breast CSCs.
Collapse
|
21
|
Zhou S, Zhu C, Pang Q, Liu HC. MicroRNA-217: A regulator of human cancer. Biomed Pharmacother 2020; 133:110943. [PMID: 33254014 DOI: 10.1016/j.biopha.2020.110943] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 10/17/2020] [Accepted: 10/25/2020] [Indexed: 12/13/2022] Open
Abstract
As highly conserved non-coding RNAs of approximately 18-24 nucleotides, microRNAs (miRNAs) regulate the expression of target genes. Multiple studies have demonstrated that miRNAs participate in the regulation of human cancer. MircoRNA-217 (miR-217) participates in the regulation of various tumors by specifically binding target genes and post-transcriptional regulation. In recent years, there have been numerous reports about miR-217 in tumor progression. MiR-217 is known mainly as a tumor suppressor, although some studies have shown that it functions as an oncomiR. Here, we review the current research related to miR-217, including its role in tumor progression and the molecular mechanisms.
Collapse
Affiliation(s)
- Shuai Zhou
- Department of Hepatobiliary Pancreatic Surgery, The First Affiliated Hospital of Bengbu Medical College, Anhui, 233000, China.
| | - Chao Zhu
- Department of Hepatobiliary Pancreatic Surgery, The First Affiliated Hospital of Bengbu Medical College, Anhui, 233000, China.
| | - Qing Pang
- Department of Hepatobiliary Pancreatic Surgery, The First Affiliated Hospital of Bengbu Medical College, Anhui, 233000, China.
| | - Hui Chun Liu
- Department of Hepatobiliary Pancreatic Surgery, The First Affiliated Hospital of Bengbu Medical College, Anhui, 233000, China.
| |
Collapse
|
22
|
Ortega MA, Fraile-Martínez O, Guijarro LG, Casanova C, Coca S, Álvarez-Mon M, Buján J, García-Honduvilla N, Asúnsolo Á. The Regulatory Role of Mitochondrial MicroRNAs (MitomiRs) in Breast Cancer: Translational Implications Present and Future. Cancers (Basel) 2020; 12:cancers12092443. [PMID: 32872155 PMCID: PMC7564393 DOI: 10.3390/cancers12092443] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 08/25/2020] [Accepted: 08/26/2020] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Mitochondrial microRNAs (mitomiRs) are an emerging field of study in a wide range of tumours including breast cancer. By targeting mitochondrial, or non-mitochondrial products, mitomiRs are able to regulate the functions of this organelle, thus controlling multiple carcinogenic processes. The knowledge of this system may provide a novel approach for targeted therapies, as potential biomarkers or helping in the diagnosis of such a complex malignancy. Abstract Breast cancer is the most prevalent and incident female neoplasm worldwide. Although survival rates have considerably improved, it is still the leading cause of cancer-related mortality in women. MicroRNAs are small non-coding RNA molecules that regulate the posttranscriptional expression of a wide variety of genes. Although it is usually located in the cytoplasm, several studies have detected a regulatory role of microRNAs in other cell compartments such as the nucleus or mitochondrion, known as “mitomiRs”. MitomiRs are essential modulators of mitochondrion tasks and their abnormal expression has been linked to the aetiology of several human diseases related to mitochondrial dysfunction, including breast cancer. This review aims to examine basic knowledge of the role of mitomiRs in breast cancer and discusses their prospects as biomarkers or therapeutic targets.
Collapse
Affiliation(s)
- Miguel A. Ortega
- Department of Medicine and Medical Specialities, Unit of Histology and Pathology, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (O.F.-M.); (C.C.); (S.C.); (M.Á.-M.); (J.B.); (N.G.-H.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain;
- Cancer Registry and Pathology Department, Hospital Universitario Principe de Asturias, 28806 Alcalá de Henares, Madrid, Spain
- Correspondence: ; Tel.: +34-91-885-4540; Fax: +34-91-885-4885
| | - Oscar Fraile-Martínez
- Department of Medicine and Medical Specialities, Unit of Histology and Pathology, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (O.F.-M.); (C.C.); (S.C.); (M.Á.-M.); (J.B.); (N.G.-H.)
| | - Luis G. Guijarro
- Department of System Biology, Unit of Biochemistry and Molecular Biology (CIBEREHD), University of Alcalá, 28801 Alcalá de Henares, Spain;
| | - Carlos Casanova
- Department of Medicine and Medical Specialities, Unit of Histology and Pathology, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (O.F.-M.); (C.C.); (S.C.); (M.Á.-M.); (J.B.); (N.G.-H.)
| | - Santiago Coca
- Department of Medicine and Medical Specialities, Unit of Histology and Pathology, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (O.F.-M.); (C.C.); (S.C.); (M.Á.-M.); (J.B.); (N.G.-H.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain;
| | - Melchor Álvarez-Mon
- Department of Medicine and Medical Specialities, Unit of Histology and Pathology, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (O.F.-M.); (C.C.); (S.C.); (M.Á.-M.); (J.B.); (N.G.-H.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain;
- Immune System Diseases-Rheumatology, Oncology Service an Internal Medicine, University Hospital Príncipe de Asturias, (CIBEREHD), 28806 Alcalá de Henares, Madrid, Spain
| | - Julia Buján
- Department of Medicine and Medical Specialities, Unit of Histology and Pathology, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (O.F.-M.); (C.C.); (S.C.); (M.Á.-M.); (J.B.); (N.G.-H.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain;
| | - Natalio García-Honduvilla
- Department of Medicine and Medical Specialities, Unit of Histology and Pathology, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (O.F.-M.); (C.C.); (S.C.); (M.Á.-M.); (J.B.); (N.G.-H.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain;
| | - Ángel Asúnsolo
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain;
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Madrid, Spain
| |
Collapse
|
23
|
Chen Z, Wang Y, Wang J, Kang M, Tang W, Chen S. Assessment of PPARGC1A, PPARGC1B, and PON1 Genetic Polymorphisms in Esophageal Squamous Cell Carcinoma Susceptibility in the Eastern Chinese Han Population: A Case-Control Study Involving 2351 Subjects. DNA Cell Biol 2020; 39:1521-1531. [PMID: 32721231 DOI: 10.1089/dna.2020.5416] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Previous studies suggested that alterations in the energy metabolism might be underlying cancer initiation and progression. Polymorphisms of genes involved in energy metabolism regulation, such as peroxisome proliferator-activated receptor gamma coactivator 1α (PPARGC1A), -β (PPARGC1B), and paraoxonase 1 (PON1), might confer susceptibility to esophageal squamous cell carcinoma (ESCC) and partially explain its pathogenesis. We investigated the effects of several single nucleotide polymorphisms (SNPs) in three metabolic-related genes (e.g., PPARGC1A, PPARGC1B, and PON1) on ESCC susceptibility. In total, 829 patients with sporadic ESCC and 1522 nontumor controls were enrolled in the study. SNPs were genotyped using PCR-ligase detection reaction. Our study revealed that the PPARGC1A rs3736265 G/A SNP significantly increased the risk for ESCC (GA vs. GG: adjusted odds ratio [OR] = 1.25, 95% confidence interval [95% CI] = 1.02-1.54, p = 0.034; GA+AA vs. GG: adjusted OR = 1.25, 95% CI = 1.03-1.52, p = 0.027]. In addition, a stratified analysis revealed that the PPARGC1A rs3736265 SNP was correlated with the development of ESCC in male and nondrinking subgroups. We also confirmed that the PPARGC1B rs17572019 G/A SNP promoted the risk of ESCC in subgroup with high alcohol intake. The PPARGC1A rs8192678 C/T polymorphism decreased the susceptibility of ESCC in men. These findings highlight that polymorphisms in PPARGC1A and PPARGC1B may contribute to ESCC susceptibility. In the future, further well-designed epidemiological studies are needed to confirm our findings.
Collapse
Affiliation(s)
- Zhan Chen
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Yafeng Wang
- Department of Cardiology, The People's Hospital of Xishuangbanna Dai Autonomous Prefecture, Jinghong, China
| | - Jusi Wang
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Mingqiang Kang
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, China.,Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China.,Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Weifeng Tang
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Shuchen Chen
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| |
Collapse
|
24
|
Lü J, Zhang C, Han J, Xu Z, Li Y, Zhen L, Zhao Q, Guo Y, Wang Z, Bischof E, Yu Z. Starvation stress attenuates the miRNA-target interaction in suppressing breast cancer cell proliferation. BMC Cancer 2020; 20:627. [PMID: 32631271 PMCID: PMC7339532 DOI: 10.1186/s12885-020-07118-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 06/26/2020] [Indexed: 11/10/2022] Open
Abstract
Background Emerging evidence has demonstrated the limited access to metabolic substrates as an effective approach to block cancer cell growth. The mechanisms remain unclear. Our previous work has revealed that miR-221/222 plays important role in regulating breast cancer development and progression through interaction with target gene p27. Results Herein, we determined the miRNA-mRNA interaction in breast cancer cells under induced stress status of starvation. Starvation stimulation attenuated the miR-221/222-p27 interaction in MDA-MB-231 cells, thereby increased p27 expression and suppressed cell proliferation. Through overexpression or knockdown of miR-221/222, we found that starvation-induced stress attenuated the negative regulation of p27 expression by miR-221/222. Similar patterns for miRNA-target mRNA interaction were observed between miR-17-5p and CyclinD1, and between mR-155 and Socs1. Expression of Ago2, one of the key components of RNA-induced silencing complex (RISC), was decreased under starvation-induced stress status, which took responsibility for the impaired miRNA-target interaction since addition of exogenous Ago2 into MDA-MB-231 cells restored the miR-221/222-p27 interaction in starvation condition. Conclusions We demonstrated the attenuated interaction between miR-221/222 and p27 by starvation-induced stress in MDA-MB-231 breast cancer cells. The findings add a new page to the general knowledge of negative regulation of gene expression by miRNAs, also demonstrate a novel mechanism through which limited access to nutrients suppresses cancer cell proliferation. These insights provide a basis for development of novel therapeutic options for breast cancer.
Collapse
Affiliation(s)
- Jinhui Lü
- Research Center for Translational Medicine, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China
| | - Chuyi Zhang
- Research Center for Translational Medicine, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China
| | - Junyi Han
- Department of Surgery, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China
| | - Zhen Xu
- Research Center for Translational Medicine, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China
| | - Yuan Li
- Research Center for Translational Medicine, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China
| | - Lixiao Zhen
- Research Center for Translational Medicine, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China
| | - Qian Zhao
- Research Center for Translational Medicine, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China
| | - Yuefan Guo
- Research Center for Translational Medicine, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China
| | - Zhaohui Wang
- Research Center for Translational Medicine, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China.,Jinzhou Medical University, Liaoning, China
| | - Evelyne Bischof
- Shanghai University of Medicine and Health Sciences Clinical Medicine Division, Shanghai, China. .,Division of Internal Medicine, University Hospital of Basel, Petersgraben 4, 4051, Basel l, Switzerland.
| | - Zuoren Yu
- Research Center for Translational Medicine, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China.
| |
Collapse
|
25
|
Amalia L, Sadeli HA, Parwati I, Rizal A, Panigoro R. Hypoxia-inducible factor-1α in acute ischemic stroke: neuroprotection for better clinical outcome. Heliyon 2020; 6:e04286. [PMID: 32637689 PMCID: PMC7327744 DOI: 10.1016/j.heliyon.2020.e04286] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 02/01/2020] [Accepted: 06/19/2020] [Indexed: 02/06/2023] Open
Abstract
Background Hypoxia-inducible factor-1α (HIF-1α) is a transcription factor which maintains cellular homeostasis in response to hypoxia. It can trigger apoptosis while stimulating angiogenesis process and decrease neurological deficit after an ischemic stroke. Up until now, this protein complex has not been widely investigated especially in stroke patient. Objective Here, we examined the potential of HIF-1α as a marker for neuroplasticity process after ischemic stroke. Methods Serum HIF-1α were measured in acute ischemic stroke patients. National Institute of Health Stroke Scale (NIHSS) were assessed on the admission and discharge day (between days 7 and 14). Ischemic stroke divided into 2 groups: large vessel disease (LVD, n = 31) and small vessel disease (SVD, n = 27). Statistical significances were calculated with Spearman rank test. Results A total of 58 patients, 31 with large artery atherosclerosis LVD and 27 with small vessel disease (SVD) were included in this study. HIF-1α level in LVD group was 0.5225 ± 0.2459 ng/mL and in SVD group was 0.3815 ± 0.121 ng/mL. HIF-1α was higher (p = 0.004) in LVD group than in SVD group. The initial NIHSS score in LVD group was 15.46 ± 2.61 and discharge NIHSS score was 13.31 ± 3.449. Initial NIHSS score in SVD group was 6.07 ± 1.82 and the discharge NIHSS was 5.703 ± 1.7055. In both SVD and LVD group, HIF-1α were significantly correlated with initial NIHSS (both p < 0.001) and discharge NIHSS (p < 0.0383 r = 0.94, p < 0.001, r = 0.93, respectively). Conclusions HIF-1α has a strong correlation with NIHSS and it may be used as predictor in acute ischemic stroke outcome.
Collapse
Affiliation(s)
- Lisda Amalia
- Department of Neurology, Faculty of Medicine, Universitas Padjadjaran, Hasan Sadikin General Hospital Bandung, Indonesia
| | - Henny Anggraini Sadeli
- Department of Neurology, Faculty of Medicine, Universitas Padjadjaran, Hasan Sadikin General Hospital Bandung, Indonesia
| | - Ida Parwati
- Department of Clinical Pathology Faculty of Medicine, Universitas Padjadjaran, Hasan Sadikin General Hospital Bandung, Indonesia
| | - Ahmad Rizal
- Department of Neurology, Faculty of Medicine, Universitas Padjadjaran, Hasan Sadikin General Hospital Bandung, Indonesia
| | - Ramdan Panigoro
- Department of Biomedical Science Faculty of Medicine, Universitas Padjadjaran, Indonesia
| |
Collapse
|
26
|
Diaz-Vegas A, Sanchez-Aguilera P, Krycer JR, Morales PE, Monsalves-Alvarez M, Cifuentes M, Rothermel BA, Lavandero S. Is Mitochondrial Dysfunction a Common Root of Noncommunicable Chronic Diseases? Endocr Rev 2020; 41:5807952. [PMID: 32179913 PMCID: PMC7255501 DOI: 10.1210/endrev/bnaa005] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Accepted: 03/12/2020] [Indexed: 12/19/2022]
Abstract
Mitochondrial damage is implicated as a major contributing factor for a number of noncommunicable chronic diseases such as cardiovascular diseases, cancer, obesity, and insulin resistance/type 2 diabetes. Here, we discuss the role of mitochondria in maintaining cellular and whole-organism homeostasis, the mechanisms that promote mitochondrial dysfunction, and the role of this phenomenon in noncommunicable chronic diseases. We also review the state of the art regarding the preclinical evidence associated with the regulation of mitochondrial function and the development of current mitochondria-targeted therapeutics to treat noncommunicable chronic diseases. Finally, we give an integrated vision of how mitochondrial damage is implicated in these metabolic diseases.
Collapse
Affiliation(s)
- Alexis Diaz-Vegas
- Advanced Center for Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina, Universidad de Chile, Santiago, Chile.,Charles Perkins Centre, School of Life and Environmental Sciences, The University of Sydney, Camperdown, Sydney, NSW, Australia
| | - Pablo Sanchez-Aguilera
- Advanced Center for Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - James R Krycer
- Charles Perkins Centre, School of Life and Environmental Sciences, The University of Sydney, Camperdown, Sydney, NSW, Australia
| | - Pablo E Morales
- Advanced Center for Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Matías Monsalves-Alvarez
- Advanced Center for Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina, Universidad de Chile, Santiago, Chile.,Institute of Nutrition and Food Technology (INTA), Universidad de Chile, Santiago, Chile
| | - Mariana Cifuentes
- Advanced Center for Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina, Universidad de Chile, Santiago, Chile.,Institute of Nutrition and Food Technology (INTA), Universidad de Chile, Santiago, Chile.,Center for Studies of Exercise, Metabolism and Cancer (CEMC), Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Beverly A Rothermel
- Department of Internal Medicine (Cardiology Division), University of Texas Southwestern Medical Center, Dallas, Texas
| | - Sergio Lavandero
- Advanced Center for Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas & Facultad de Medicina, Universidad de Chile, Santiago, Chile.,Department of Internal Medicine (Cardiology Division), University of Texas Southwestern Medical Center, Dallas, Texas.,Center for Studies of Exercise, Metabolism and Cancer (CEMC), Facultad de Medicina, Universidad de Chile, Santiago, Chile
| |
Collapse
|
27
|
Bouguerra H, Amal G, Clavel S, Boussen H, Louet JF, Gati A. Leptin decreases BC cell susceptibility to NK lysis via PGC1A pathway. Endocr Connect 2020; 9:578-586. [PMID: 32449691 PMCID: PMC7354724 DOI: 10.1530/ec-20-0109] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 05/21/2020] [Indexed: 12/12/2022]
Abstract
Large prospective studies established a link between obesity and breast cancer (BC) development. Yet, the mechanisms underlying this association are not fully understood. Among the diverse adipocytokine secreted by hypertrophic adipose tissue, leptin is emerging as a key candidate molecule linking obesity and cancer, since it promotes proliferation and invasiveness of tumors. However, the potential implication of leptin on tumor escape mechanisms remains unknown. This study aims to explore the effect of leptin on tumor resistance to NK lysis and the underlying mechanism. We found that leptin promotes both BC resistance to NK92-mediated lysis and β oxidation on MCF-7, by the up-regulation of a master regulator of mitochondrial biogenesis, the peroxisome proliferator activated receptor coactivator-1 α (PGC1A). Using adenoviral approaches, we show that acute elevation of PGC1A enhances the fatty acid oxidation pathway and decreases the susceptibility of BC cells to NK92-mediated lysis. Importantly, we identified the involvement of PGC1A and leptin in the regulation of hypoxia inducible factor-1 alpha (HIF1A) expression by tumor cells. We further demonstrate that basal BC cells MDA-MB-231 and BT-20 exhibit an increased PGC1A mRNA level and an enhanced oxidative phosphorylation activity; in comparison with luminal BC cells MCF7 and MDA-361, which are associated with more resistance NK92 lysis. Altogether, our results demonstrate for the first time how leptin could promote tumor resistance to immune attacks. Reagents blocking leptin or PGC1A activity might aid in developing new therapeutic strategies to limit tumor development in obese BC patients.
Collapse
Affiliation(s)
- Hichem Bouguerra
- Université Tunis El-Manar, Faculté des Sciences de Tunis, Laboratoire de Génétique, Immunologie et pathologies Humaines, Tunis, Tunisie
- Université Côte d'Azur, INSERM, C3M, Team Cellular and Molecular Physiopathology of Obesity and Diabetes, Nice, France
| | - Gorrab Amal
- Université Tunis El-Manar, Faculté des Sciences de Tunis, Laboratoire de Génétique, Immunologie et pathologies Humaines, Tunis, Tunisie
| | - Stephan Clavel
- Université Côte d'Azur, INSERM, C3M, Team Cellular and Molecular Physiopathology of Obesity and Diabetes, Nice, France
| | - Hamouda Boussen
- Département d’Oncologie Médicale, Hôpital Abderrahman Mami, Ariana, Tunisia
| | - Jean-François Louet
- Université Côte d'Azur, INSERM, C3M, Team Cellular and Molecular Physiopathology of Obesity and Diabetes, Nice, France
| | - Asma Gati
- Université Tunis El-Manar, Faculté des Sciences de Tunis, Laboratoire de Génétique, Immunologie et pathologies Humaines, Tunis, Tunisie
- Correspondence should be addressed to A Gati:
| |
Collapse
|
28
|
Calahorra J, Martínez-Lara E, Granadino-Roldán JM, Martí JM, Cañuelo A, Blanco S, Oliver FJ, Siles E. Crosstalk between hydroxytyrosol, a major olive oil phenol, and HIF-1 in MCF-7 breast cancer cells. Sci Rep 2020; 10:6361. [PMID: 32286485 PMCID: PMC7156391 DOI: 10.1038/s41598-020-63417-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 03/30/2020] [Indexed: 12/14/2022] Open
Abstract
Olive oil intake has been linked with a lower incidence of breast cancer. Hypoxic microenvironment in solid tumors, such as breast cancer, is known to play a crucial role in cancer progression and in the failure of anticancer treatments. HIF-1 is the foremost effector in hypoxic response, and given that hydroxytyrosol (HT) is one of the main bioactive compounds in olive oil, in this study we deepen into its modulatory role on HIF-1. Our results in MCF-7 breast cancer cells demonstrate that HT decreases HIF-1α protein, probably by downregulating oxidative stress and by inhibiting the PI3K/Akt/mTOR pathway. Strikingly, the expression of HIF-1 target genes does not show a parallel decrease. Particularly, adrenomedullin and vascular endothelial growth factor are up-regulated by high concentrations of HT even in HIF-1α silenced cells, pointing to HIF-1-independent mechanisms of regulation. In fact, we show, by in silico modelling and transcriptional analysis, that high doses of HT may act as an agonist of the aryl hydrocarbon receptor favoring the induction of these angiogenic genes. In conclusion, we suggest that the effect of HT in a hypoxic environment is largely affected by its concentration and involves both HIF-1 dependent and independent mechanisms.
Collapse
Affiliation(s)
- Jesús Calahorra
- Departamento de Biología Experimental, Universidad de Jaén, Campus Las Lagunillas s/n, Jaén, 23071, Spain
| | - Esther Martínez-Lara
- Departamento de Biología Experimental, Universidad de Jaén, Campus Las Lagunillas s/n, Jaén, 23071, Spain
| | - José M Granadino-Roldán
- Departamento de Química Física y Analítica, Universidad de Jaén, Campus Las Lagunillas s/n, Jaén, 23071, Spain
| | - Juan M Martí
- Instituto López Neyra de Parasitología y Biomedicina, IPBLN, CSIC PTS-Granada, Armilla, 18016, Spain
| | - Ana Cañuelo
- Departamento de Biología Experimental, Universidad de Jaén, Campus Las Lagunillas s/n, Jaén, 23071, Spain
| | - Santos Blanco
- Departamento de Biología Experimental, Universidad de Jaén, Campus Las Lagunillas s/n, Jaén, 23071, Spain
| | - F Javier Oliver
- Instituto López Neyra de Parasitología y Biomedicina, IPBLN, CSIC PTS-Granada, Armilla, 18016, Spain
| | - Eva Siles
- Departamento de Biología Experimental, Universidad de Jaén, Campus Las Lagunillas s/n, Jaén, 23071, Spain.
| |
Collapse
|
29
|
Clinicopathological and prognostic value of hypoxia-inducible factor-1α in breast cancer: a meta-analysis including 5177 patients. Clin Transl Oncol 2020; 22:1892-1906. [PMID: 32166713 DOI: 10.1007/s12094-020-02332-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 02/25/2020] [Indexed: 12/12/2022]
Abstract
PURPOSE Mounting studies have investigated the clinicopathological and prognostic value of hypoxia-inducible factor-1α (HIF-1α) in breast cancer (BC), yet conclusions remain controversial. Therefore, we conducted this meta-analysis to clarify this issue. METHODS All relevant studies were searched using Cochrane Library, Web of Science, PubMed, and EMBASE online databases. Pooled odds ratios (ORs) and hazard ratios (HRs) with 95% confidence intervals (CIs) were applied to evaluate the clinicopathological and prognostic value of HIF-1α, respectively. Subgroup analysis and sensitivity analysis were performed to investigate heterogeneity and stability of the results. Begg's funnel plot and Egger's test were used to examine publication bias. RESULTS A total of 31 eligible studies including 5177 subjects were enrolled. Of these, 25 studies assessed the prognostic role of HIF-1α and included 4546 individuals. Twenty-three studies involving 3277 individuals evaluated the clinicopathological significance of HIF-1α. High expression level of HIF-1α was correlated with poor overall survival (OS) (HR = 1.59, 95% CI = 1.40-1.80, P < 0.001), disease-free survival (DFS) (HR = 1.87, 95% CI = 1.53-2.28, P < 0.001), relapse-free survival (HR = 1.36, 95% CI = 1.07-1.73, P = 0.001), and cancer-specific survival (HR = 1.55, 95% CI = 1.10-2.19, P = 0.012). Pooled data from studies using multivariate survival analysis also showed that HIF-1α expression was associated with worse OS (HR = 1.59, 95% CI = 1.32-1.92, P < 0.001) and DFS (HR = 1.60, 95% CI = 1.39-1.84, P < 0.001). Additionally, high HIF-1α expression was associated with advanced tumor-node-metastasis stage, positive lymph-node status, negative ER status, ductal type, advanced histologic grade, high Ki67 expression, and strong VEGF expression. CONCLUSION HIF-1α might serve as an independent prognostic biomarker and a promising therapeutic target for BC. Future large-scale prospective randomized trials are needed to confirm our findings.
Collapse
|
30
|
Wan J, Ling X, Rao Z, Peng B, Ding G. Independent prognostic value of HIF-1α expression in radiofrequency ablation of lung cancer. Oncol Lett 2020; 19:849-857. [PMID: 31897199 PMCID: PMC6924154 DOI: 10.3892/ol.2019.11130] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 09/20/2019] [Indexed: 01/11/2023] Open
Abstract
Radiofrequency ablation (RFA) is widely used in the treatment of lung cancer. Hypoxia-inducible factor-1α (HIF-1α) is a crucial transcription factor regulating oxygen homeostasis that is involved in tumor cell metastasis. The present study investigated the impact of HIF-1α expression and other factors, such as postoperative blood CD4+/CD8+ ratio, on the prognosis of patients with lung cancer who had received RFA treatment. A total of 80 patients with lung cancer were recruited between January 2011 and October 2016 at The Shenzhen People's Hospital. Lung cancer was confirmed following pathological or histological examination. All patients underwent RFA treatment. Patients were followed up for 6–66 months. HIF-1α expression in lung cancer tissues was assessed by immunohistochemistry. Multivariate survival analysis was performed using Cox proportional hazards model. The results demonstrated that HIF-1α level was low in 36 patients and overexpressed in 44 patients with lung cancer. Kaplan-Meier (KM) curve analysis demonstrated that the overall survival time of patients with high HIF-1α expression was significantly shorter compared with patients with low HIF-1α expression (P<0.05). Furthermore, the results from the KM model and log-rank test revealed that age, Union for International Cancer Control stage, primary or metastatic cancer, chemotherapy, postoperative blood CD4+/CD8+ ratio, Eastern Cooperative Oncology Group performance status and HIF-1α expression had significant effects on overall survival of patients with lung cancer. The results from Cox analysis demonstrated that high HIF-1α expression, advanced age, clinical staging and chemotherapy were independent risk factors for the prognosis of lung cancer following RFA treatment, and that high HIF-1α expression was associated with the increased risk (5.91-fold) of mortality. In conclusion, the present study demonstrated that HIF-1α expression was increased in lung cancer tissues and was associated with the prognosis of patients with lung cancer who were treated with RFA. These findings suggest that HIF-1α expression may be considered as a marker for evaluating the prognosis of these patients.
Collapse
Affiliation(s)
- Jun Wan
- Department of Thoracic Surgery, The Shenzhen People's Hospital, The Second Clinical Medicine College of Jinan University, Shenzhen, Guangdong 518020, P.R. China
| | - Xiean Ling
- Department of Thoracic Surgery, The Shenzhen People's Hospital, The Second Clinical Medicine College of Jinan University, Shenzhen, Guangdong 518020, P.R. China
| | - Zhanpeng Rao
- Department of Thoracic Surgery, The Shenzhen People's Hospital, The Second Clinical Medicine College of Jinan University, Shenzhen, Guangdong 518020, P.R. China
| | - Bin Peng
- Department of Thoracic Surgery, The Shenzhen People's Hospital, The Second Clinical Medicine College of Jinan University, Shenzhen, Guangdong 518020, P.R. China
| | - Guanggui Ding
- Department of Thoracic Surgery, The Shenzhen People's Hospital, The Second Clinical Medicine College of Jinan University, Shenzhen, Guangdong 518020, P.R. China
| |
Collapse
|
31
|
Wu LF, Xu GP, Zhao Q, Zhou LJ, Wang D, Chen WX. The association between hypoxia inducible factor 1 subunit alpha gene rs2057482 polymorphism and cancer risk: a meta-analysis. BMC Cancer 2019; 19:1123. [PMID: 31744467 PMCID: PMC6862742 DOI: 10.1186/s12885-019-6329-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Accepted: 11/04/2019] [Indexed: 02/06/2023] Open
Abstract
Background The rs2057482 polymorphism in the hypoxia inducible factor 1 subunit alpha (HIF1A) gene has been reported to be associated with a risk of several types of cancer, but this association has not yet been definitively confirmed. We performed this meta-analysis to determine whether rs2057482 is associated with overall cancer risk. Methods The PubMed, Embase, and Web of Science databases were searched for the potential studies about the association between the rs2057482 and cancer risk. The data of genotype frequencies in cases with cancer and controls were extracted from the selected studies. Odds ratios (ORs) and the corresponding 95% confidence intervals (CIs) were calculated to determine the strength of the associations. Results The meta-analysis showed an association between the rs2057482 polymorphism and overall cancer risk. However, a stratified analysis of ethnicity did not show any significant association between rs2057482 and cancer risk in the Asian population. Conclusions The rs2057482 polymorphism was associated with decreased overall cancer risk, based on the currently available studies. However, this conclusion needs verification by further well-designed epidemiology studies that examine different cancer types and more subjects.
Collapse
Affiliation(s)
- Li-Fang Wu
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Gui-Ping Xu
- Transfusion Department, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qing Zhao
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Li-Jing Zhou
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Ding Wang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Wei-Xian Chen
- Department of Laboratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China.
| |
Collapse
|
32
|
Kim H, Kim HS, Moon WK. Comparison of transcriptome expression alterations by chronic exposure to low-dose bisphenol A in different subtypes of breast cancer cells. Toxicol Appl Pharmacol 2019; 385:114814. [PMID: 31715268 DOI: 10.1016/j.taap.2019.114814] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 10/22/2019] [Accepted: 11/08/2019] [Indexed: 12/25/2022]
Abstract
The impacts of chronic bisphenol A (BPA) exposure suspected to be a potential risk factor for breast cancer progression are not thoroughly understood in different subtypes of breast cancer cells (BCCs). This study aimed to compare the differentially expressed genes (DEGs) and biological functions in MCF-7 (luminal A), SK-BR3 (HER2-enriched) and MDA-MB-231 (triple-negative) cells exposed to BPA at an environmentally human-relevant low dose (10-8 M) for 30 days, by using the approach of RNA sequencing and online informatics tools. BPA-exposure resulted in 172, 137, and 139 DEGs in MCF-7/BPA, SK-BR3/BPA, and MDA-MB-231/BPA, respectively. The significantly enriched gene ontology terms of DEGs in each cell were different: cellular response to gonadotropin-releasing hormone, negative regulation of fibrinolysis, choline metabolism, glutamate signaling pathways and coagulation pathway in MCF-7/BPA; positive regulation of inflammatory response and VEGF/VEGFR signaling pathways in SK-BR3/BPA; negative regulation of keratinocyte proliferation and HIF signaling pathways in MDA-MB-231/BPA cells. The immune network analysis of DEGs across the breast cancer cells indicated NKT, NK and T cell activation and dendritic cell migration by regulating the expression of immunomodulatory genes. High expression of IL19, CA9 and SPARC identified in MCF-7/BPA, SK-BR3/BPA, and MDA-MB-231/BPA are detrimental gene signatures to predict poor overall survival in luminal A, HER2-enriched and triple-negative breast cancer patients, respectively. These findings indicate chronic BPA exposure has dissimilar impacts on the regulation of gene expression and diverse biological functions, including immune modulation, in different subtypes of BCCs.
Collapse
Affiliation(s)
- Hyelim Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea
| | - Hoe Suk Kim
- Department of Radiology, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea
| | - Woo Kyung Moon
- Department of Radiology, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea; Department of Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea; Department of Radiology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea.
| |
Collapse
|
33
|
Romero-Garcia S, Prado-Garcia H, Valencia-Camargo AD, Alvarez-Pulido A. Lactic Acidosis Promotes Mitochondrial Biogenesis in Lung Adenocarcinoma Cells, Supporting Proliferation Under Normoxia or Survival Under Hypoxia. Front Oncol 2019; 9:1053. [PMID: 31681589 PMCID: PMC6811519 DOI: 10.3389/fonc.2019.01053] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 09/27/2019] [Indexed: 11/17/2022] Open
Abstract
Lactic acidosis, glucose deprivation and hypoxia are conditions frequently found in solid tumors because, among other reasons, tumors switch to Warburg effect and secrete high levels of lactate, which decreases the pH (<6. 9) in the microenvironment. We hypothesized that lung cancer cells consume lactate and induce mitochondrial biogenesis to support survival and proliferation in lactic acidosis with glucose deprivation even under hypoxia. We examined lung adenocarcinoma cell lines (A-427 and A-549), a breast cancer cell line (MCF-7) and non-transformed fibroblasts (MRC-5). Cells were cultured using RPMI-1640 medium with 28 mM lactate varying pH (6.2 or 7.2) under normoxia (atmospheric O2) or hypoxia (2% O2). Cellular growth was followed during 96 h, as well as lactate, glutamine and glutamate levels, which were measured using a biochemical analyzer. The expression levels of monocarboxylate transporters (MCT1 and MCT4) were evaluated by flow cytometry. To evaluate mitochondrial biogenesis, mitochondrial mass was analyzed by flow cytometry and epifluorescence microscopy. Also, mitochondrial DNA (mtDNA) was measured by qPCR. Transcript levels of Nuclear Respiratory Factors (NRF-1 and NRF-2) and Transcription Factor A Mitochondrial (TFAM) were determined using RT-qPCR. The specific growth rate of A-549 and A-427 cells increased in lactic acidosis compared with neutral lactosis, either under normoxia or hypoxia, a phenomenon that was not observed in MRC-5 fibroblasts. Under hypoxia, A-427 and MCF-7 cells did not survive in neutral lactosis but survived in lactic acidosis. Under lactic acidosis, A-427 and MCF-7 cells increased MCT1 levels, reduced MCT4 levels and consumed higher lactate amounts, while A-549 cells consumed glutamine and decreased MCT1 and MCT4 levels with respect to neutral lactosis condition. Lactic acidosis, either under normoxia or hypoxia, increased mitochondrial mass and mtDNA levels compared with neutral lactosis in all tumor cells but not in fibroblasts. A-549 and MCF-7 cells increased levels of NRF-1, NRF-2, and TFAM with respect to MRC-5 cells, whereas A-427 cells upregulated these transcripts under lactic acidosis compared with neutral lactosis. Thus, lung adenocarcinoma cells induce mitochondrial biogenesis to support survival and proliferation in lactic acidosis with glucose deprivation.
Collapse
|
34
|
Fuggetta MP, Spanu P, Ulgheri F, Deligia F, Carta P, Mannu A, Trotta V, De Cicco R, Barra A, Zona E, Morelli F. A New Synthetic Spiroketal: Studies on Antitumor Activity on Murine Melanoma Model In Vivo and Mechanism of Action In Vitro. Anticancer Agents Med Chem 2019; 19:567-578. [PMID: 30706794 DOI: 10.2174/1871520619666190131141400] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 11/05/2018] [Accepted: 01/19/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND In a previous study, we synthesised a new spiroketal derivative, inspired to natural products, that has shown high antiproliferative activity, potent telomerase inhibition and proapoptotic activity on several human cell lines. OBJECTIVE This work focused on the study of in vivo antitumor effect of this synthetic spiroketal on a murine melanoma model. In order to shed additional light on the origin of the antitumor effect, in vitro studies were performed. METHODS Spiroketal was administered to B16F10 melanoma mice at a dose of 5 mg/Kg body weight via intraperitoneum at alternate days for 15 days. Tumor volume measures were made every 2 days starting after 12 days from cells injection. The effects of the spiroketal on tumor growth inhibition, apoptosis induction, and cell cycle modification were investigated in vitro on B16 cells. HIF1α gene expression, the inhibition of cells migration and the changes induced in cytoskeleton conformation were evaluated. RESULTS Spiroketal displayed proapoptotic activity and high antitumor activity in B16 cells with nanomolar IC50. Moreover it has shown to inhibit cell migration, to strongly reduce the HIF1α expression and to induce strongly deterioration of cytoskeleton structure. A potent dose-dependent antitumor efficacy in syngenic B16/C57BL/6J murine model of melanoma was observed with the suppression of tumor growth by an average of 90% at a dose of 5 mg/kg. CONCLUSION The synthesized spiroketal shows high antitumor activity in the B16 cells in vitro at nM concentration and a dose-dependent antitumor efficacy in syngenic B16/C57BL/6J mice. The results suggest that this natural product inspired spiroketal may have a potential application in melanoma therapy.
Collapse
Affiliation(s)
- Maria P Fuggetta
- Istituto di Farmacologia Traslazionale-Consiglio Nazionale delle Ricerche, Roma, Italy
| | - Pietro Spanu
- Istituto di Chimica Biomolecolare- Consiglio Nazionale delle Ricerche, Sassari, Italy
| | - Fausta Ulgheri
- Istituto di Chimica Biomolecolare- Consiglio Nazionale delle Ricerche, Sassari, Italy
| | - Francesco Deligia
- Istituto di Chimica Biomolecolare- Consiglio Nazionale delle Ricerche, Sassari, Italy
| | - Paola Carta
- Istituto di Chimica Biomolecolare- Consiglio Nazionale delle Ricerche, Sassari, Italy
| | - Alberto Mannu
- Istituto di Chimica Biomolecolare- Consiglio Nazionale delle Ricerche, Sassari, Italy
| | - Veronica Trotta
- Istituto di Genetica e Biofisica A. Buzzati Traverso-Consiglio Nazionale delle Ricerche, Napoli, Italy
| | - Rosanna De Cicco
- Istituto di Genetica e Biofisica A. Buzzati Traverso-Consiglio Nazionale delle Ricerche, Napoli, Italy
| | - Adriano Barra
- Istituto di Genetica e Biofisica A. Buzzati Traverso-Consiglio Nazionale delle Ricerche, Napoli, Italy
| | - Enrica Zona
- Istituto di Genetica e Biofisica A. Buzzati Traverso-Consiglio Nazionale delle Ricerche, Napoli, Italy
| | - Franco Morelli
- Istituto di Genetica e Biofisica A. Buzzati Traverso-Consiglio Nazionale delle Ricerche, Napoli, Italy
| |
Collapse
|
35
|
Chen W, Cheng X, Wang X, Wang J, Wen X, Xie C, Liao C. Clinical implications of hypoxia-inducible factor-1α and caveolin-1 overexpression in isocitrate dehydrogenase-wild type glioblastoma multiforme. Oncol Lett 2019; 17:2867-2873. [PMID: 30854062 PMCID: PMC6365898 DOI: 10.3892/ol.2019.9929] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 12/06/2018] [Indexed: 12/18/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most common type of primary brain tumour in adults, and presents a very low survival rate. Isocitrate dehydrogenase (IDH)1/2 mutations have been found in ~12% of glioblastomas and are associated with long-term GBM survival. However, the risk factors that influence the prognosis of IDH-wild type GBM remain unclear. Hypoxia-inducible factor (HIF)-1α, an important oxygen-regulated transcription factor, has been demonstrated to serve a crucial role in tumour development and to be associated with a poor prognosis. In addition, caveolin-1 (CAV1) is a plasma membrane organizing protein, the expression of which can also be regulated by a hypoxic microenvironment. The present study therefore aimed to examine the expression levels of HIF-1α and CAV1, and their association with GBM prognosis. Reverse transcription-quantitative polymerase chain reaction and western blotting were performed to analyse the expression levels of HIF-1α and CAV1 in paired GBM tumour and adjacent non-tumour tissues. Immunohistochemistry was used to analyse the expression of the two proteins in paraffin-embedded tissues obtained from 42 patients with IDH-wild type GBM. Statistical analyses were performed to examine the correlation between HIF-1α and CAV1 expression and patient prognosis. The results revealed hat the expression levels of HIF-1α and CAV1 were upregulated in IDH-wild type GBM tissues compared to their paired non-tumour tissues (P<0.001). The expression of CAV1 was significantly correlated with high HIF-1α expression (P<0.01). In addition, overexpression of HIF-1α and CAV1 was markedly associated with a poor prognosis (P<0.001). In conclusion, HIF-1α and CAV1 may represent potential biomarkers for IDH-wild type GBM prognosis and potential targets for the development of therapies extending GBM survival.
Collapse
Affiliation(s)
- Wenli Chen
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Xing Cheng
- Department of Spine Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Xiaobo Wang
- Department of Spine Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Jinshan Wang
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Xiaoling Wen
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Chaofan Xie
- Department of Spine Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Chuangxin Liao
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong 510080, P.R. China
| |
Collapse
|
36
|
Nie C, Lv H, Bie L, Hou H, Chen X. Hypoxia-inducible factor 1-alpha expression correlates with response to neoadjuvant chemotherapy in women with breast cancer. Medicine (Baltimore) 2018; 97:e13551. [PMID: 30572455 PMCID: PMC6319826 DOI: 10.1097/md.0000000000013551] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Hypoxia-inducible factor 1-alpha (HIF-1a) has been shown to contribute to resistance to chemotherapy in breast cancer. The purpose of this study was to investigate whether HIF-1a is predictive for pathological response and the prognostic value of HIF-1a in local advanced breast undergoing neoadjuvant chemotherapy.Two hundred twenty patients with none-metastatic locally advanced invasive breast cancer (stages II-III) that subsequently received neoadjuvant chemotherapy were included in an observational study to assess the HIF-1a protein expression by immunohistochemistry. Associations between HIF-1a expression and pathological complete response (pCR) were analyzed using univariate and multivariate analysis. Independent prognostic factors for RFS were identified by multivariate Cox's proportional hazard analysis. A P value < .05 was considered to be statistically significant.The median age was 46 years, Luminal A, Luminal B, HER2-positive, and triple-negative accounted for 3.6%, 57.7%, 7.0% and 16.0%, respectively. A total of 41 patients (18.6%) achieved a pCR after neoadjuvant chemotherapy in the present study. HIF-1α negative patients had a significantly higher pCR rate than HIF-1α positive patients (P = .027). Multivariate analysis demonstrated that HIF-1α negative expression is an independent favorable predictor of pCR. Multivariate Cox regression analysis demonstrated that the HIF-1a expression before NCT showed an independent prognostic value for RFS (HR = 4.168, 95% CI: 1.012-17.170, P = .048).HIF-1a expression correlates with pCR in breast cancer undergoing neoadjuvant chemotherapy. Absent expression of HIF-1a was associated with a better pathological response and could indicate a favorable prognosis in non-pCR breast cancer patients.
Collapse
Affiliation(s)
- Caiyun Nie
- Department of Oncology, The Affiliated Cancer Hospital of Zhengzhou University
- He’nan Province Tumor Hospital, Zhengzhou, China
| | - Huifang Lv
- Department of Oncology, The Affiliated Cancer Hospital of Zhengzhou University
- He’nan Province Tumor Hospital, Zhengzhou, China
| | - Liangyu Bie
- Department of Oncology, The Affiliated Cancer Hospital of Zhengzhou University
- He’nan Province Tumor Hospital, Zhengzhou, China
| | - Honglin Hou
- Department of Oncology, The Affiliated Cancer Hospital of Zhengzhou University
- He’nan Province Tumor Hospital, Zhengzhou, China
| | - Xiaobing Chen
- Department of Oncology, The Affiliated Cancer Hospital of Zhengzhou University
- He’nan Province Tumor Hospital, Zhengzhou, China
| |
Collapse
|
37
|
PGC1α: Friend or Foe in Cancer? Genes (Basel) 2018; 9:genes9010048. [PMID: 29361779 PMCID: PMC5793199 DOI: 10.3390/genes9010048] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 01/15/2018] [Accepted: 01/16/2018] [Indexed: 12/14/2022] Open
Abstract
The PGC1 family (Peroxisome proliferator-activated receptor γ (PPARγ) coactivators) of transcriptional coactivators are considered master regulators of mitochondrial biogenesis and function. The PGC1α isoform is expressed especially in metabolically active tissues, such as the liver, kidneys and brain, and responds to energy-demanding situations. Given the altered and highly adaptable metabolism of tumor cells, it is of interest to investigate PGC1α in cancer. Both high and low levels of PGC1α expression have been reported to be associated with cancer and worse prognosis, and PGC1α has been attributed with oncogenic as well as tumor suppressive features. Early in carcinogenesis PGC1α may be downregulated due to a protective anticancer role, and low levels likely reflect a glycolytic phenotype. We suggest mechanisms of PGC1α downregulation and how these might be connected to the increased cancer risk that obesity is now known to entail. Later in tumor progression PGC1α is often upregulated and is reported to contribute to increased lipid and fatty acid metabolism and/or a tumor cell phenotype with an overall metabolic plasticity that likely supports drug resistance as well as metastasis. We conclude that in cancer PGC1α is neither friend nor foe, but rather the obedient servant reacting to metabolic and environmental cues to benefit the tumor cell.
Collapse
|
38
|
Pan T, Wang Q, Zhu L, Qi J, You T, Han Y. Downregulation of hypoxia-inducible factor-1α contributes to impaired megakaryopoiesis in immune thrombocytopenia. Thromb Haemost 2017; 117:1875-1886. [DOI: 10.1160/th17-03-0155] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 06/23/2017] [Indexed: 01/15/2023]
Abstract
SummaryImpaired megakaryocyte maturation and exaggerated platelet destruction play a pivotal role in the pathogenesis of immune thrombocytopenia (ITP). Previous studies have shown that HIF-1α promotes the homing and engraftment of haematopoietic stem cells (HSCs), thereby stimulating HSC differentiation. However, whether HIF-1α plays a role in megakaryocytic maturation and platelet destruction in ITP remains elusive. Using enzyme-linked immunosorbent assays (ELISAs), we demonstrated that there were lower HIF-1α levels in the bone marrow (BM) of ITP patients than in that of healthy donors and patients with chemotherapy-related thrombocytopenia. Subjects with lower megakaryocyte (<100/slide) and platelet (<30 × 109/L) counts exhibited significantly decreased BM HIF-1α levels, compared to those with higher megakaryocyte (≥100/slide) and platelet (≥30 × 109/L) counts. To test whether HIF-1α regulates megakaryopoiesis and platelet production, megakaryocytes derived from mouse BM cells were treated with an HIF-1α activator (IOX-2; 50 µM) or inhibitor (PX-478; 50 µM). PX-478 significantly decreased HIF-1α expression, cell size, and the populations of CD41-positive and high-ploidy cells. Importantly, to evaluate the role of HIF-1α as a potential therapeutic target in ITP, mouse BM cells were incubated with plasma from ITP patients in the presence or absence of IOX-2. IOX-2 significantly attenuated the ITP plasma-induced decrease in cell size as well as the proportions of CD41-positive and high-ploidy cells. In addition, IOX-2 increased the number of megakaryocytes from mouse BM cells treated with ITP plasma. Our findings indicate that decreased HIF-1α may contribute to impaired megakaryopoiesis in ITP, and HIF-1α may provide a potential therapy for ITP patients.Supplementary Material to this article is available online at www.thrombosis-online.com.
Collapse
|
39
|
Bialesova L, Xu L, Gustafsson JÅ, Haldosen LA, Zhao C, Dahlman-Wright K. Estrogen receptor β2 induces proliferation and invasiveness of triple negative breast cancer cells: association with regulation of PHD3 and HIF-1α. Oncotarget 2017; 8:76622-76633. [PMID: 29100336 PMCID: PMC5652730 DOI: 10.18632/oncotarget.20635] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 08/17/2017] [Indexed: 12/25/2022] Open
Abstract
The two estrogen receptor (ER) subtypes, ERα and ERβ, belong to the nuclear receptor superfamily. The human ERβ variant ERβ2 is proposed to be expressed at higher levels than ERβ1 in many breast tumors and it has been suggested that ERβ2, in contrast to ERβ1, is associated with aggressive phenotypes of various cancers. However, the role of endogenous ERβ2 in breast cancer cells remains elusive. In this study, we identified that triple negative breast cancer (TNBC) cell lines express endogenous ERβ2, but not ERα or ERβ1. This allows novel studies of endogenous ERβ2 functions independent of ERα and ERβ1. We show that overexpression of ERβ2 in TNBC cells increased whereas knockdown of endogenous ERβ2 decreased cell proliferation and cell invasion. To elucidate the molecular mechanism responsible for these cellular phenotypes, we assayed ERβ2 dependent global gene expression profiles. We show that ERβ2 decreases prolyl hydroxylase 3 (PHD3) gene expression and further show that this is associated with increased hypoxia inducible factor 1α (HIF-1α) protein levels, thus providing a possible mechanism for the invasive phenotype. These results are further supported by analysing the expression of ERβ2 and PHD3 in breast tumor samples where a negative correlation between ERβ2 and PHD3 expression was observed. Together, we demonstrate that ERβ2 has an important role in enhancing cell proliferation and invasion, beyond modulation of ERβ and ERβ1 signalling which might contribute to the invasive characteristics of TNBC. The invasive phenotype could potentially be mediated through transcriptional repression of PHD3 and increased HIF-1α protein levels.
Collapse
Affiliation(s)
- Lucia Bialesova
- Department of Biosciences and Nutrition, Novum, Karolinska Institutet, Huddinge S-141 83, Sweden
| | - Li Xu
- Department of Biosciences and Nutrition, Novum, Karolinska Institutet, Huddinge S-141 83, Sweden
| | - Jan-Åke Gustafsson
- Department of Biosciences and Nutrition, Novum, Karolinska Institutet, Huddinge S-141 83, Sweden.,Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX 77204-5056, USA
| | - Lars-Arne Haldosen
- Department of Biosciences and Nutrition, Novum, Karolinska Institutet, Huddinge S-141 83, Sweden
| | - Chunyan Zhao
- Department of Biosciences and Nutrition, Novum, Karolinska Institutet, Huddinge S-141 83, Sweden
| | - Karin Dahlman-Wright
- Department of Biosciences and Nutrition, Novum, Karolinska Institutet, Huddinge S-141 83, Sweden
| |
Collapse
|
40
|
Wang XX, Sun HY, Yang QZ, Guo B, Sai Y, Zhang J. Hypoxia-inducible factor-1α and glucose transporter 1 in the malignant transformation of oral lichen planus. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2017; 10:8369-8376. [PMID: 31966688 PMCID: PMC6965416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Accepted: 03/18/2017] [Indexed: 06/10/2023]
Abstract
Hypoxia-inducible factor-1α (HIF-1α) and glucose transporter 1 (GLUT1) are key factors in numerous physiological and pathological processes. However, studies on their involvement in the pathogenesis of oral lichen planus (OLP) and its progression toward oral squamous cell carcinomas (OSCC) are scarce. In this study, we examined the protein and gene expressions of both HIF-1α and GLUT1 in normal mucosa, nonatrophic OLP (OLPI), atrophic OLP (OLPII), and OSCC resulting from OLP. Tissues were obtained from 60 cases of OLP patients (n=36 for OLPI, n=24 for OLPII), 20 cases of OSCC patients and 30 healthy control individuals. In addition, in order to investigate if the pathological changes are due to hypoxia, we cultured keratinocytes under hypoxia conditions and measured the expression of HIF-1α and GLUT1. The results indicated that the expressions of HIF-1α and GLUT1 were gradually amplified from normal mucosa to OLPI, OLPII, and OSCC. The expression of both HIF-1α and GLUT1 in OLPII was significantly greater than OLPII. Likewise, the HIF-1α and GLUT1 expressions in OSCC were markedly higher compared to both OLPI and OLPII. Similar trends were obtained in real time PCR and Western blot analyses. A progressive increased micro-vessel density (MVD) was also recorded from normal mucosa to OLPI, OLPII, and OSCC. Moreover, the correlation analysis revealed significant positive correlations between HIF-1α and GLUT1 which were both correlated with MVD in the OLP and OSCC groups. Culture of keratinocytes isolated from OLP tissues under hypoxic and normoxic conditions showed a time-dependent inhibition of keratinocyte proliferation and increased expression of HIF-1α and GLUT1 under hypoxia conditions. In summary, we provided new evidence that hypoxia markers HIF-1α and GLUT1 are upregulated in OLP and are potentially involved in pathological changes leading to malignant transformation of OLP. Further characterization of these factors will provide new ideas for the diagnosis and treatment of OLP.
Collapse
Affiliation(s)
- Xia-Xia Wang
- Department of Stomatology, Huashan Hospital, Fudan UniversityShanghai, P. R. China
| | - Hong-Ying Sun
- Department of Stomatology, Huashan Hospital, Fudan UniversityShanghai, P. R. China
| | - Qiao-Zhen Yang
- Department of Stomatology, Huashan Hospital, Fudan UniversityShanghai, P. R. China
| | - Bin Guo
- School of Life Science, Institute of GeneticsShanghai, P. R. China
| | - Yin Sai
- School of Life Science, Institute of GeneticsShanghai, P. R. China
| | - Jie Zhang
- Department of Stomatology, Huashan Hospital, Fudan UniversityShanghai, P. R. China
| |
Collapse
|
41
|
Kim B, Jung JW, Jung J, Han Y, Suh DH, Kim HS, Dhanasekaran DN, Song YS. PGC1α induced by reactive oxygen species contributes to chemoresistance of ovarian cancer cells. Oncotarget 2017; 8:60299-60311. [PMID: 28947972 PMCID: PMC5601140 DOI: 10.18632/oncotarget.19140] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 06/10/2017] [Indexed: 12/13/2022] Open
Abstract
Malignant cells are subjected to high levels of oxidative stress that arise from the increased production of reactive oxygen species (ROS) due to their altered metabolism. They activate antioxidant mechanisms to relieve the oxidative stress, and thereby acquire resistance to chemotherapeutic agents. In the present study, we found that PGC1α, a key molecule that both increases mitochondrial biogenesis and activates antioxidant enzymes, enhances chemoresistance in response to ROS generated by exposure of cells to ovarian sphere-forming culture conditions. Cells in the cultured spheres exhibited stem cell-like characteristics, and maintained higher ROS levels than their parent cells. Intriguingly, scavenging ROS diminished the aldehyde dehydrogenase (ALDH)-positive cell population, and inhibited proliferation of the spheres. ROS production triggered PGC1α expression, which in turn caused changes to mitochondrial biogenesis and activity within the spheres. The drug-resistant phenotype was observed in both spheres and PGC1α-overexpressing parent cells, and conversely, PGC1α knockdown sensitized the spheres to cisplatin treatment. Similarly, floating malignant cells derived from patient ascitic fluid included an ALDH-positive population and exhibited the tendency of a positive correlation between expressions of multidrug resistance protein 1 (MDR1) and PGC1α. The present study suggests that ROS-induced PGC1α mediates chemoresistance, and represents a novel therapeutic target to overcome chemoresistance in ovarian cancer.
Collapse
Affiliation(s)
- Boyun Kim
- Cancer Research Institute, College of Medicine, Seoul National University, Seoul 03080, Korea.,Nano System Institute, Seoul National University, Seoul 08826, Korea
| | - Je Won Jung
- Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea
| | - Jaeyoung Jung
- Cancer Research Institute, College of Medicine, Seoul National University, Seoul 03080, Korea.,WCU Biomodulation, Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Korea
| | - Youngjin Han
- Cancer Research Institute, College of Medicine, Seoul National University, Seoul 03080, Korea.,WCU Biomodulation, Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Korea
| | - Dong Hoon Suh
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seongnam 13620, Korea
| | - Hee Seung Kim
- Department of Obstetrics and Gynecology, College of Medicine, Seoul National University, Seoul 03080, Korea
| | - Danny N Dhanasekaran
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73012, United States of America
| | - Yong Sang Song
- Cancer Research Institute, College of Medicine, Seoul National University, Seoul 03080, Korea.,WCU Biomodulation, Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Korea.,Department of Obstetrics and Gynecology, College of Medicine, Seoul National University, Seoul 03080, Korea
| |
Collapse
|
42
|
Wang S, Ren T, Huang Y, Bao X, Sun K, Shen D, Guo W. BMPR2 and HIF1- α overexpression in resected osteosarcoma correlates with distant metastasis and patient survival. Chin J Cancer Res 2017; 29:447-454. [PMID: 29142464 DOI: 10.21147/j.issn.1000-9604.2017.05.09] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Objective Bone morphogenetic protein receptor 2 (BMPR2) and hypoxia-inducible factor 1-α (HIF1-α) existed abnormal expression in several types of cancer. However, their expressions and related roles in osteosarcoma are largely unknown. Methods To investigate the clinical significance of BMPR2 and HIF1-α in osteosarcoma, we analyzed their expression levels in 103 osteosarcoma specimens by immunochemistry. Meanwhile, we conducted a follow-up to examine the metastatic behavior and overall survival (OS) of osteosarcoma patients. Results Among 103 tissues, 61 cases had BMPR2-positive expression and 57 cases had HIF1-α positive expression. A significant correlation was noticed between BMPR2 and HIF1-α expression in osteosarcoma specimens (P=0.035). Receiver-operating characteristic (ROC) curves were calculated to investigate the predictive value of the two markers in tumor metastasis. By means of univariate and multivariate analysis, BMPR2 and HIF1-α expression, as well as higher tumor grade, were identified as significant risk factors for OS in patients with osteosarcoma. Kaplan-Meier survival analysis revealed that the patients with BMPR2 and HIF1-α positive expression had worse OS compared with patients with BMPR2-negative or HIF1-α-negative staining. Conclusions It can be concluded that BMPR2 and HIF1-α expression is highly correlated with metastatic behavior in patients with osteosarcoma and can serve as predictive markers for metastasis and OS of these patients.
Collapse
Affiliation(s)
- Shidong Wang
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing 100044, China.,Beijing Key Laboratory of Musculoskeletal Tumor, Beijing 100044, China
| | - Tingting Ren
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing 100044, China.,Beijing Key Laboratory of Musculoskeletal Tumor, Beijing 100044, China
| | - Yi Huang
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing 100044, China.,Beijing Key Laboratory of Musculoskeletal Tumor, Beijing 100044, China
| | - Xing Bao
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing 100044, China.,Beijing Key Laboratory of Musculoskeletal Tumor, Beijing 100044, China
| | - Kunkun Sun
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing 100044, China
| | - Danhua Shen
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing 100044, China
| | - Wei Guo
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing 100044, China.,Beijing Key Laboratory of Musculoskeletal Tumor, Beijing 100044, China
| |
Collapse
|
43
|
Cheng CF, Ku HC, Lin H. Functional alpha 1 protease inhibitor produced by a human hepatoma cell line. ACTA ACUST UNITED AC 1982; 19:ijms19113447. [PMID: 30400212 PMCID: PMC6274980 DOI: 10.3390/ijms19113447] [Citation(s) in RCA: 284] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 10/23/2018] [Accepted: 10/30/2018] [Indexed: 12/13/2022]
Abstract
Alpha 1 protease inhibitor antigen was identified in the culture medium of the human ascites hepatoma cell line SK-HEP-1. Trypsin inhibitory activity and alpha 1 Pl antigen accumulated in serum-free medium concomitantly over a period of several days. Radioactive alpha 1 Pl antigen was detected in conditioned medium from cultures supplemented with 35S-L-methionine, indicating a synthesis and release of the protein. Alpha 1 Pl antigen in conditioned medium appeared to be antigenically identical to that in human plasma, and the newly synthesized (radiolabeled) antigen co-migrated with plasma, alpha 1 Pl after immunoelectrophoresis or SDS-polyacrylamide gel electrophoresis. Moreover, evidence is presented that the synthesized inhibitor exhibits functional activity, since the 35S-labeled alpha 1 Pl in conditioned medium complexes with trypsin. We conclude that SK-HEP-1 cells in culture produce functionally active alpha 1 Pl which may be identical to that in plasma.
Collapse
Affiliation(s)
- Ching-Feng Cheng
- Department of Pediatrics, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 23142, Taiwan.
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan.
- Department of Pediatrics, Tzu Chi University, Hualien 97004, Taiwan.
| | - Hui-Chen Ku
- Department of Pediatrics, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 23142, Taiwan.
| | - Heng Lin
- Institute of Pharmacology, Taipei Medical University, 250 Wu-Hsing Street, Taipei 11031, Taiwan.
- Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
| |
Collapse
|