1
|
Liu W, Li H, Gao Y, Zhang X, Wei Z, Yang D, Jin M, Qiu Z, Shen Z, Chen Z, Qiao Y, Pu L, Yan C, Zhang S, Wang X, Li J. Mitochondrial dysfunction of peripheral blood mononuclear cells is associated with lung carcinogenesis. Int Immunopharmacol 2024; 133:111958. [PMID: 38608441 DOI: 10.1016/j.intimp.2024.111958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 03/19/2024] [Accepted: 03/26/2024] [Indexed: 04/14/2024]
Abstract
The composition, quantity, and function of peripheral blood mononuclear cells (PBMCs) are closely correlated with tumorigenesis. However, the mechanisms of PBMCs in lung cancer are not clear. Mitochondria are energy factories of cells, and almost all cellular functions rely on their energy metabolism level. The present study aimed to test whether the mitochondrial function of PBMCs directly determines their tumor immune monitoring function. We recruited 211 subjects, including 105 healthy controls and 106 patients with recently diagnosed with lung cancer. The model of lung carcinogenesis induced by BaP was used in animal experiment, and the Bap carcinogenic metabolite, Benzo(a)pyren-7,8-dihydrodiol-9,10-epoxide (BPDE), was used in cell experiment. We found that mitochondrial function of PBMCs decreased significantly in patients with new lung cancer, regardless of age. In vivo, BaP caused PBMC mitochondrial dysfunction in mice before the appearance of visible malignant tissue. Moreover, mitochondrial function decreased significantly in mice with lung cancers induced by BaP compared to those without lung cancer after BaP intervention. In vitro, BPDE also induced mitochondrial dysfunction and reduced the aggressiveness of PBMCs toward cancer cells. Furthermore, the changes in mitochondrial energy metabolism gene expression caused by BPDE are involved in this process. Thus, the mitochondrial function of PBMCs is a potential prognostic biomarker or therapeutic target to improve clinical outcomes in patients with lung cancer.
Collapse
Affiliation(s)
- Weili Liu
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China.
| | - Hua Li
- Department of Endoscopy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Yuan Gao
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China; Maternity&Child Care Center of Dezhou, Shandong, China
| | - Xuelian Zhang
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Zilin Wei
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Dong Yang
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Min Jin
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Zhigang Qiu
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Zhiqiang Shen
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Zhaoli Chen
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Yamei Qiao
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Lingling Pu
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Changqing Yan
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Shuang Zhang
- Tianjin Centers for Disease Control and Prevention, Tianjin 300011, China
| | - Xinxing Wang
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China.
| | - Junwen Li
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China.
| |
Collapse
|
2
|
Osanloo M, Pishamad S, Ghanbariasad A, Zarenezhad E, Alipanah M, Alipanah H. Comparison effects of Ferula gummosa essential oil and Beta-pinene Alginate nanoparticles on human melanoma and breast cancer cells proliferation and apoptotic index in short term normobaric hyperoxic model. BMC Complement Med Ther 2023; 23:428. [PMID: 38017466 PMCID: PMC10683214 DOI: 10.1186/s12906-023-04266-4] [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: 08/08/2023] [Accepted: 11/18/2023] [Indexed: 11/30/2023] Open
Abstract
BACKGROUND Breast cancer is the most common cancer among women, and melanoma is the most dreadful type of skin cancer. Due to the side effects of chemotherapy drugs, the development of new herbal nano-medicines has been considered. METHODS This study first investigated the chemical composition of Ferula gummosa essential oil using GC-MS analysis; β-pinene, with 61.57%, was the major compound. Next, alginate nanoparticles containing β-pinene and the essential oil with particle sizes of 174 ± 7 and 137 ± 6 nm were prepared. Meanwhile, their zeta potentials were 12.4 ± 0.7 and 28.1 ± 1 mV. Besides, the successful loading of β-pinene and the essential oil in nanoparticles was confirmed using ATR-FTIR analysis. After that, their effects on viability and apoptotic index of human melanoma and breast cancer cells were investigated in normoxia and normobaric hyperoxia (NBO) conditions. RESULTS The best efficacy on A-375 and MDA-MB-231 cells was achieved by alginate nanoparticles containing the EO at hyperoxic and normoxia conditions; IC50 76 and 104 µg/mL. Besides, it affected apoptosis-involved genes; as Bax/Bcl-2 ratio was higher than 1, conditions for induction of apoptosis were obtained. Higher sensitivity was observed in the A-375 cell line treated with Alg-EO in the NBO model. CONCLUSIONS Alginate nanoparticles containing F. gummosa EO could be considered for further investigation in anticancer studies. Also, it may be expected that NBO can be a new strategy for delaying cancer progression and improving nanotherapy efficacy.
Collapse
Affiliation(s)
- Mahmoud Osanloo
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Somayyeh Pishamad
- Student Research Committee, Fasa University of Medical Sciences, Fasa, Iran
| | - Ali Ghanbariasad
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Elham Zarenezhad
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Media Alipanah
- Faculty of Veterinary Medicine, Razi University, Kermanshah, Iran
| | - Hiva Alipanah
- Department of Physiology, School of Medicine, Fasa University of Medical Sciences, Fasa, Iran.
| |
Collapse
|
3
|
Lee H, Kim IK, Im J, Jin BS, Kim HH, Kim SW, Yeo CD, Lee SH. Effects of aging on accompanying intermittent hypoxia in a bleomycin-induced pulmonary fibrosis mouse model. Korean J Intern Med 2023; 38:934-944. [PMID: 37793985 PMCID: PMC10636542 DOI: 10.3904/kjim.2023.090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 05/26/2023] [Accepted: 05/30/2023] [Indexed: 10/06/2023] Open
Abstract
BACKGROUND/AIMS Obstructive sleep apnea (OSA) is prevalent in older patients with idiopathic pulmonary fibrosis (IPF); however, it is underrecognized. OSA is characterized by intermittent hypoxia (IH) and sleep fragmentation. In this study, we evaluated the effects of IH in an older mouse model of bleomycin-induced lung fibrosis. METHODS Bleomycin-induced mice (C57BL/6, female) were randomly divided into four groups of young vs. old and room air (RA)-exposed vs. IH-exposed. Mice were exposed to RA or IH (20 cycles/h, FiO2 nadir 7 ± 0.5%, 8 h/day) for four weeks. The mice were sacrificed on day 28, and blood, bronchoalveolar lavage (BAL) fluid, and lung tissue samples were obtained. RESULTS The bleomycin-induced IH-exposed (EBI) older group showed more severe inflammation, fibrosis, and oxidative stress than the other groups. The levels of inflammatory cytokines in the serum and BAL fluid increased in the EBI group. Hydroxyproline levels in the lung tissue increased markedly in the EBI group. CONCLUSION This study demonstrates the possible harmful impact of OSA in an elderly mouse model of lung fibrosis. This study further suggests that older patients with IPF and OSA may be more of a concern than younger patients with IPF. Further research is required in this area.
Collapse
Affiliation(s)
- Heayon Lee
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Eunpyeong St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - In Kyoung Kim
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Eunpyeong St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jeonghyeon Im
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Eunpyeong St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Bae Suk Jin
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Eunpyeong St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Hwan Hee Kim
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Eunpyeong St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Sei Won Kim
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Eunpyeong St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Chang Dong Yeo
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Eunpyeong St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Sang Haak Lee
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Eunpyeong St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
4
|
Zhuang Y, Liu K, He Q, Gu X, Jiang C, Wu J. Hypoxia signaling in cancer: Implications for therapeutic interventions. MedComm (Beijing) 2023; 4:e203. [PMID: 36703877 PMCID: PMC9870816 DOI: 10.1002/mco2.203] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 12/14/2022] [Accepted: 12/18/2022] [Indexed: 01/25/2023] Open
Abstract
Hypoxia is a persistent physiological feature of many different solid tumors and a key driver of malignancy, and in recent years, it has been recognized as an important target for cancer therapy. Hypoxia occurs in the majority of solid tumors due to a poor vascular oxygen supply that is not sufficient to meet the needs of rapidly proliferating cancer cells. A hypoxic tumor microenvironment (TME) can reduce the effectiveness of other tumor therapies, such as radiotherapy, chemotherapy, and immunotherapy. In this review, we discuss the critical role of hypoxia in tumor development, including tumor metabolism, tumor immunity, and tumor angiogenesis. The treatment methods for hypoxic TME are summarized, including hypoxia-targeted therapy and improving oxygenation by alleviating tumor hypoxia itself. Hyperoxia therapy can be used to improve tissue oxygen partial pressure and relieve tumor hypoxia. We focus on the underlying mechanisms of hyperoxia and their impact on current cancer therapies and discuss the prospects of hyperoxia therapy in cancer treatment.
Collapse
Affiliation(s)
- Yan Zhuang
- State Key Laboratory of Pharmaceutical BiotechnologyNational Institute of Healthcare Data Science at Nanjing UniversityJiangsu Key Laboratory of Molecular MedicineMedicineMedical School of Nanjing UniversityNanjing UniversityNanjingChina
| | - Kua Liu
- State Key Laboratory of Pharmaceutical BiotechnologyNational Institute of Healthcare Data Science at Nanjing UniversityJiangsu Key Laboratory of Molecular MedicineMedicineMedical School of Nanjing UniversityNanjing UniversityNanjingChina
| | - Qinyu He
- State Key Laboratory of Pharmaceutical BiotechnologyNational Institute of Healthcare Data Science at Nanjing UniversityJiangsu Key Laboratory of Molecular MedicineMedicineMedical School of Nanjing UniversityNanjing UniversityNanjingChina
| | - Xiaosong Gu
- Microecological, Regenerative and Microfabrication Technical Platform for Biomedicine and Tissue EngineeringJinan Microecological Biomedicine Shandong LaboratoryJinan CityChina
| | - Chunping Jiang
- State Key Laboratory of Pharmaceutical BiotechnologyNational Institute of Healthcare Data Science at Nanjing UniversityJiangsu Key Laboratory of Molecular MedicineMedicineMedical School of Nanjing UniversityNanjing UniversityNanjingChina
- Microecological, Regenerative and Microfabrication Technical Platform for Biomedicine and Tissue EngineeringJinan Microecological Biomedicine Shandong LaboratoryJinan CityChina
| | - Junhua Wu
- State Key Laboratory of Pharmaceutical BiotechnologyNational Institute of Healthcare Data Science at Nanjing UniversityJiangsu Key Laboratory of Molecular MedicineMedicineMedical School of Nanjing UniversityNanjing UniversityNanjingChina
- Microecological, Regenerative and Microfabrication Technical Platform for Biomedicine and Tissue EngineeringJinan Microecological Biomedicine Shandong LaboratoryJinan CityChina
| |
Collapse
|
5
|
Salvagno M, Coppalini G, Taccone FS, Strapazzon G, Mrakic-Sposta S, Rocco M, Khalife M, Balestra C. The Normobaric Oxygen Paradox-Hyperoxic Hypoxic Paradox: A Novel Expedient Strategy in Hematopoiesis Clinical Issues. Int J Mol Sci 2022; 24:ijms24010082. [PMID: 36613522 PMCID: PMC9820104 DOI: 10.3390/ijms24010082] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/17/2022] [Accepted: 12/18/2022] [Indexed: 12/24/2022] Open
Abstract
Hypoxia, even at non-lethal levels, is one of the most stressful events for all aerobic organisms as it significantly affects a wide spectrum of physiological functions and energy production. Aerobic organisms activate countless molecular responses directed to respond at cellular, tissue, organ, and whole-body levels to cope with oxygen shortage allowing survival, including enhanced neo-angiogenesis and systemic oxygen delivery. The benefits of hypoxia may be evoked without its detrimental consequences by exploiting the so-called normobaric oxygen paradox. The intermittent shift between hyperoxic-normoxic exposure, in addition to being safe and feasible, has been shown to enhance erythropoietin production and raise hemoglobin levels with numerous different potential applications in many fields of therapy as a new strategy for surgical preconditioning aimed at frail patients and prevention of postoperative anemia. This narrative review summarizes the physiological processes behind the proposed normobaric oxygen paradox, focusing on the latest scientific evidence and the potential applications for this strategy. Future possibilities for hyperoxic-normoxic exposure therapy include implementation as a synergistic strategy to improve a patient's pre-surgical condition, a stimulating treatment in critically ill patients, preconditioning of athletes during physical preparation, and, in combination with surgery and conventional chemotherapy, to improve patients' outcomes and quality of life.
Collapse
Affiliation(s)
- Michele Salvagno
- Department of Intensive Care, Hôpital Universitaire de Bruxelles (HUB), 1070 Brussels, Belgium
| | - Giacomo Coppalini
- Department of Intensive Care, Hôpital Universitaire de Bruxelles (HUB), 1070 Brussels, Belgium
| | - Fabio Silvio Taccone
- Department of Intensive Care, Hôpital Universitaire de Bruxelles (HUB), 1070 Brussels, Belgium
| | - Giacomo Strapazzon
- Institute of Mountain Emergency Medicine, Eurac Research, 39100 Bolzano, Italy
| | - Simona Mrakic-Sposta
- Institute of Clinical Physiology—National Research Council (CNR-IFC), 20162 Milano, Italy
| | - Monica Rocco
- Dipartimento di Scienze Medico Chirurgiche e Medicina Traslazionale, Sapienza University of Rome, 00189 Rome, Italy
| | - Maher Khalife
- Department of Anesthesiology, Institut Jules Bordet, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
| | - Costantino Balestra
- Environmental, Occupational, Aging (Integrative) Physiology Laboratory, Haute Ecole Bruxelles-Brabant (HE2B), 1050 Brussels, Belgium
- Anatomical Research and Clinical Studies, Vrije Universiteit Brussels (VUB), 1090 Brussels, Belgium
- DAN Europe Research Division (Roseto-Brussels), 1020 Brussels, Belgium
- Physical Activity Teaching Unit, Motor Sciences Department, Université Libre de Bruxelles (ULB), 1050 Brussels, Belgium
- Correspondence:
| |
Collapse
|
6
|
Liu X, Qin H, Li Z, Lv Y, Feng S, Zhuang W, Quan X, Guo C, Chen C, Zhang H. Inspiratory hyperoxia suppresses lung cancer metastasis through a MYC/SLC1A5-dependent metabolic pathway. Eur Respir J 2022; 60:13993003.00062-2022. [PMID: 35680143 PMCID: PMC9712851 DOI: 10.1183/13993003.00062-2022] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 05/18/2022] [Indexed: 12/14/2022]
Abstract
The lack of knowledge about the effect of inspiratory hyperoxia on the lung-specific tumour microenvironment and progression of lung cancer has attracted considerable attention. This study proposes that inspiratory hyperoxia has special significance for the malignant phenotype of lung cancer cells. The effects of different oxygenation parameters on the proliferation, apoptosis, invasion and migration of lung cancer cells were systematically evaluated in vitro and in vivo Our results reveal that inspiratory hyperoxia treatment (60% oxygen, 6 h·day-1) not only has no tumour progression-promoting effects, but also suppresses lung cancer metastasis and promotes long-term survival. In addition, we combined transcriptome, proteome and metabolome analysis and found that hyperoxia treatment induced significant intracellular metabolic changes in lung cancer cells. Overall, we established that MYC/SLC1A5-induced metabolic reprogramming and glutamine addiction is a new mechanism that drives lung cancer metastasis, which can be significantly suppressed by inspiratory hyperoxia treatment. These findings are relevant to the debate on the perils, promises and antitumour effect of inspiratory hyperoxia, especially for patients with lung cancer.
Collapse
Affiliation(s)
- Xiucheng Liu
- Thoracic Surgery Laboratory, Xuzhou Medical University, Xuzhou, China,Department of Thoracic Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China,Shanghai Engineering Research Center of Lung Transplantation, Shanghai, China,Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China,Xiucheng Liu, Hao Qin and Zheng Li contributed equally to this work
| | - Hao Qin
- Department of Thoracic Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China,Xiucheng Liu, Hao Qin and Zheng Li contributed equally to this work
| | - Zheng Li
- Department of Thoracic Surgery, Huadong Hospital Affiliated to FuDan University, Shanghai, China,Xiucheng Liu, Hao Qin and Zheng Li contributed equally to this work
| | - Yin Lv
- Department of Thoracic Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Shoujie Feng
- Department of Thoracic Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Wei Zhuang
- Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Xiaoyu Quan
- Department of Thoracic Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Chen Guo
- Department of Thoracic Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Chang Chen
- Shanghai Engineering Research Center of Lung Transplantation, Shanghai, China,Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China,Hao Zhang and Chang Chen contributed equally to this article as lead authors and co-corresponding authors
| | - Hao Zhang
- Thoracic Surgery Laboratory, Xuzhou Medical University, Xuzhou, China,Department of Thoracic Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China,Hao Zhang and Chang Chen contributed equally to this article as lead authors and co-corresponding authors,Corresponding author: Hao Zhang ()
| |
Collapse
|
7
|
Eymin B. Inspiratory hyperoxia: a new way to prevent metastasis through metabolism reprogramming in non-small cell lung cancer. Eur Respir J 2022; 60:60/6/2201357. [PMID: 36455962 DOI: 10.1183/13993003.01357-2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 09/01/2022] [Indexed: 12/03/2022]
Affiliation(s)
- Beatrice Eymin
- Univ. Grenoble Alpes, INSERM 1209, CNRS 5309, Institute For Advanced Biosciences, Team RNA Splicing, Cell Signaling and Response to Therapies, Grenoble, France
| |
Collapse
|
8
|
Im J, Kwon HY, Kim IK, Yeo CD, Kim SW, Lee H, Kang HS, Lee SH. Normobaric hyperoxia re-sensitizes paclitaxel-resistant lung cancer cells. Mol Cell Toxicol 2022. [DOI: 10.1007/s13273-022-00225-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
9
|
Khalaf MM, Abo-Youssef AM, Malak MN, Hamzawy MA. Novel therapeutic modalities target cell signaling of Renin-Angiotensin system/NF-κB-induced cell cycle arrest and apoptosis in urethane-induced lung cancer in mice: An in vivo study. J Biochem Mol Toxicol 2022; 36:e23162. [PMID: 35822566 DOI: 10.1002/jbt.23162] [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: 12/12/2021] [Revised: 05/20/2022] [Accepted: 07/01/2022] [Indexed: 11/07/2022]
Abstract
BACKGROUND Lung cancer has risen to the top of the list of cancer-related deaths worldwide. Aliskiren is a direct renin inhibitor. AIM This study aims to investigate the impact of cell signaling of Renin-Angiotensin system (RAS)/NF-κB on lung cancer by investigating the potential therapeutic effects of aliskiren for lung cancer treatment in urethane-induced lung cancer in mice. METHODS Male BALB/c mice were randomly assigned to one of five treatment groups for 150 days, including (1) normal control; (2) aliskiren (25 mg/kg/i.p) daily, (3) urethane at a dose of 1.5 g/kg (i.p) at Day 1 and 60 (nonsmall cell lung cancer[NSCLC] group) (4) NSCLC mice received carboplatin (15 mg/kg/i.p) every other day for the last 4 successive weeks and (5) NSCLC mice treated with aliskiren daily. Tumor size was determined based on blood sampling, and lungs were isolated for biochemical analysis, western blot analysis assay, and histopathological examination. RESULTS Urethane demonstrated significant changes in all biochemical and molecular parameters and histological patterns. Aliskiren-treated mice had significantly lower levels of NF-κB p65, Bcl-2, cyclin D1, ICAM-1, MMP-2, and Nrf2, with an increase in the catalytic activity of caspase-3 due to its RAS inhibitory mechanism. The combined urethane administration with aliskiren demonstrated a significant improvement in the histopathological examination. CONCLUSION RAS/NF-B cell signaling is a potential therapeutic target for preventing and treating lung adenocarcinoma, evidenced by the fundamental cytotoxic mechanism and attenuation of metastasis and angiogenesis induced by the treatment of NSCLC mice with aliskiren.
Collapse
Affiliation(s)
- Marwa M Khalaf
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Amira M Abo-Youssef
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Marina N Malak
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Mohamed A Hamzawy
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Fayoum University, Fayoum, Egypt
| |
Collapse
|
10
|
Alpuim Costa D, Sampaio-Alves M, Netto E, Fernandez G, Oliveira E, Teixeira A, Daniel PM, Bernardo GS, Amaro C. Hyperbaric Oxygen Therapy as a Complementary Treatment in Glioblastoma-A Scoping Review. Front Neurol 2022; 13:886603. [PMID: 35847231 PMCID: PMC9283648 DOI: 10.3389/fneur.2022.886603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 05/24/2022] [Indexed: 11/13/2022] Open
Abstract
Glioblastoma (GBM) is the most common and aggressive malignant brain tumor in adults. The mainstay of management for GBM is surgical resection, radiation (RT), and chemotherapy (CT). Even with optimized multimodal treatment, GBM has a high recurrence and poor survival rates ranging from 12 to 24 months in most patients. Recently, relevant advances in understanding GBM pathophysiology have opened new avenues for therapies for recurrent and newly diagnosed diseases. GBM's hypoxic microenvironment has been shown to be highly associated with aggressive biology and resistance to RT and CT. Hyperbaric oxygen therapy (HBOT) may increase anticancer therapy sensitivity by increasing oxygen tension within the hypoxic regions of the neoplastic tissue. Previous data have investigated HBOT in combination with cytostatic compounds, with an improvement of neoplastic tissue oxygenation, inhibition of HIF-1α activity, and a significant reduction in the proliferation of GBM cells. The biological effect of ionizing radiation has been reported to be higher when it is delivered under well-oxygenated rather than anoxic conditions. Several hypoxia-targeting strategies reported that HBOT showed the most significant effect that could potentially improve RT outcomes, with higher response rates and survival and no serious adverse events. However, further prospective and randomized studies are necessary to validate HBOT's effectiveness in the 'real world' GBM clinical practice.
Collapse
Affiliation(s)
- Diogo Alpuim Costa
- Haematology and Oncology Department, CUF Oncologia, Lisbon, Portugal
- NOVA Medical School (NMS), Faculdade de Ciências Médicas (FCM), Lisbon, Portugal
- Faculty of Medicine, University of Lisbon, Lisbon, Portugal
- Centro de Medicina Subaquática e Hiperbárica, Azinhaga dos Ulmeiros, Lisbon, Portugal
- Centro Hiperbárico de Cascais, Cascais, Portugal
| | - Mafalda Sampaio-Alves
- Faculty of Medicine, University of Porto, Oporto, Portugal
- PTSurg – Portuguese Surgical Research Collaborative, Lisbon, Portugal
| | - Eduardo Netto
- Radioncology Department, Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), E.P.E., Lisbon, Portugal
| | | | - Edson Oliveira
- Faculty of Medicine, University of Lisbon, Lisbon, Portugal
- Neurosurgery Department, Cluster CUF Descobertas, Lisbon, Portugal
| | - Andreia Teixeira
- Faculty of Medicine, University of Lisbon, Lisbon, Portugal
- Centro de Medicina Subaquática e Hiperbárica, Azinhaga dos Ulmeiros, Lisbon, Portugal
| | - Pedro Modas Daniel
- Centro de Medicina Subaquática e Hiperbárica, Azinhaga dos Ulmeiros, Lisbon, Portugal
| | - Guilherme Silva Bernardo
- Faculty of Medicine, University of Lisbon, Lisbon, Portugal
- Centro de Medicina Subaquática e Hiperbárica, Azinhaga dos Ulmeiros, Lisbon, Portugal
- Urology Department, Hospital Professor Doutor Fernando Fonseca, Amadora, Portugal
| | - Carla Amaro
- Centro de Medicina Subaquática e Hiperbárica, Azinhaga dos Ulmeiros, Lisbon, Portugal
- Otorhinolaryngology Department, CUF Descobertas, Lisbon, Portugal
| |
Collapse
|
11
|
Herrera-Campos AB, Zamudio-Martinez E, Delgado-Bellido D, Fernández-Cortés M, Montuenga LM, Oliver FJ, Garcia-Diaz A. Implications of Hyperoxia over the Tumor Microenvironment: An Overview Highlighting the Importance of the Immune System. Cancers (Basel) 2022; 14:2740. [PMID: 35681719 PMCID: PMC9179641 DOI: 10.3390/cancers14112740] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 05/26/2022] [Accepted: 05/30/2022] [Indexed: 02/04/2023] Open
Abstract
Hyperoxia is used in order to counteract hypoxia effects in the TME (tumor microenvironment), which are described to boost the malignant tumor phenotype and poor prognosis. The reduction of tumor hypoxic state through the formation of a non-aberrant vasculature or an increase in the toxicity of the therapeutic agent improves the efficacy of therapies such as chemotherapy. Radiotherapy efficacy has also improved, where apoptotic mechanisms seem to be implicated. Moreover, hyperoxia increases the antitumor immunity through diverse pathways, leading to an immunopermissive TME. Although hyperoxia is an approved treatment for preventing and treating hypoxemia, it has harmful side-effects. Prolonged exposure to high oxygen levels may cause acute lung injury, characterized by an exacerbated immune response, and the destruction of the alveolar-capillary barrier. Furthermore, under this situation, the high concentration of ROS may cause toxicity that will lead not only to cell death but also to an increase in chemoattractant and proinflammatory cytokine secretion. This would end in a lung leukocyte recruitment and, therefore, lung damage. Moreover, unregulated inflammation causes different consequences promoting tumor development and metastasis. This process is known as protumor inflammation, where different cell types and molecules are implicated; for instance, IL-1β has been described as a key cytokine. Although current results show benefits over cancer therapies using hyperoxia, further studies need to be conducted, not only to improve tumor regression, but also to prevent its collateral damage.
Collapse
Affiliation(s)
- Ana Belén Herrera-Campos
- Instituto de Parasitología y Biomedicina López Neyra, CSIC, 18016 Granada, Spain; (A.B.H.-C.); (E.Z.-M.); (D.D.-B.); (M.F.-C.)
| | - Esteban Zamudio-Martinez
- Instituto de Parasitología y Biomedicina López Neyra, CSIC, 18016 Granada, Spain; (A.B.H.-C.); (E.Z.-M.); (D.D.-B.); (M.F.-C.)
- Consorcio de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain;
| | - Daniel Delgado-Bellido
- Instituto de Parasitología y Biomedicina López Neyra, CSIC, 18016 Granada, Spain; (A.B.H.-C.); (E.Z.-M.); (D.D.-B.); (M.F.-C.)
- Consorcio de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain;
| | - Mónica Fernández-Cortés
- Instituto de Parasitología y Biomedicina López Neyra, CSIC, 18016 Granada, Spain; (A.B.H.-C.); (E.Z.-M.); (D.D.-B.); (M.F.-C.)
- Consorcio de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain;
| | - Luis M. Montuenga
- Consorcio de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain;
- Program in Solid Tumors, CIMA-University of Navarra, 31008 Pamplona, Spain
- Navarra Health Research Institute (IDISNA), 31008 Pamplona, Spain
| | - F. Javier Oliver
- Instituto de Parasitología y Biomedicina López Neyra, CSIC, 18016 Granada, Spain; (A.B.H.-C.); (E.Z.-M.); (D.D.-B.); (M.F.-C.)
- Consorcio de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain;
| | - Angel Garcia-Diaz
- Instituto de Parasitología y Biomedicina López Neyra, CSIC, 18016 Granada, Spain; (A.B.H.-C.); (E.Z.-M.); (D.D.-B.); (M.F.-C.)
- Consorcio de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain;
| |
Collapse
|
12
|
AboYoussef AM, Khalaf MM, Malak MN, Hamzawy MA. Repurposing of sildenafil as antitumour; induction of cyclic guanosine monophosphate/protein kinase G pathway, caspase-dependent apoptosis and pivotal reduction of Nuclear factor kappa light chain enhancer of activated B cells in lung cancer. J Pharm Pharmacol 2021; 73:1080-1091. [PMID: 33856030 DOI: 10.1093/jpp/rgab049] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 02/23/2021] [Indexed: 12/20/2022]
Abstract
OBJECTIVES Lung cancer is one of the most frequent types of cancers that lead to death. Sildenafil is a potent inhibitor of phosphodiesterase-5 and showed potential anticancer effects, which has not yet been fully evaluated. Thus, this study aims to investigate the potential anticancer effect of sildenafil in urethane-induced lung cancer in BALB/c mice. METHODS Five-week-old male BALB/c mice were treated with either (i) normal saline only, (ii) sildenafil only 50 mg kg-1/ P.O every other day for the last four successive weeks, (iii) urethane 1.5 gm kg-1 i.p (at day 1 and day 60), (iv) carboplatin after urethane induction, or (v) sildenafil after urethane induction. KEY FINDINGS It was shown that sildenafil significantly increased the levels of cGMP and Caspase-3 with a reduction of NF-κB, Bcl-2, Cyclin D1, intercellular adhesion molecule 1, matrix metalloproteinase-2 levels and normalisation of Nrf2 along with pronounced improvement in the histological patterns. CONCLUSIONS These results indicated that sildenafil markedly induces cell cycle arrest, apoptosis and inhibits the metastatic activity through activation of cyclic guanosine monophosphate/protein kinase G pathway and down-regulation of cyclin D1 and nuclear factor kappa light chain enhancer of activated B cells with downstream anti-apoptotic gene Bcl-2, which underscores the critical importance of future using sildenafil in the treatment of lung cancer.
Collapse
Affiliation(s)
- Amira M AboYoussef
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Marwa M Khalaf
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Marina N Malak
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Mohamed A Hamzawy
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Fayoum University, Fayoum, Egypt
| |
Collapse
|
13
|
Abstract
IMPACT STATEMENT Tumor hypoxia promotes cancer cell aggressiveness, and is strongly associated with poor prognosis across multiple tumor types. The hypoxic microenvironments inside tumors also limit the effectiveness of radiotherapy, chemotherapy, and immunotherapy. Several approaches to eliminate hypoxic state in tumors have been proposed to delay cancer progression and improve therapeutic efficacies. This review will summarize current knowledge on hyperoxia, used alone or in combination with other therapeutic modalities, in cancer treatment. Molecular mechanisms and undesired side effects of hyperoxia will also be discussed.
Collapse
Affiliation(s)
- Sei W Kim
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Eunpyeong St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 03312, Republic of Korea
| | - In K Kim
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Eunpyeong St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 03312, Republic of Korea
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Sang H Lee
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Eunpyeong St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 03312, Republic of Korea
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| |
Collapse
|
14
|
Hyperoxia Alters Ultrastructure and Induces Apoptosis in Leukemia Cell Lines. Biomolecules 2020; 10:biom10020282. [PMID: 32059539 PMCID: PMC7072400 DOI: 10.3390/biom10020282] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 01/29/2020] [Accepted: 02/11/2020] [Indexed: 12/11/2022] Open
Abstract
Oxygenation conditions are crucial for growth and tumor progression. Recent data suggests a decrease in cancer cell proliferation occurring after exposure to normobaric hyperoxia. Those changes are associated with fractal dimension. The purpose of this research was to study the impact of hyperoxia on apoptosis and morphology of leukemia cell lines. Two hematopoietic lymphoid cancer cell lines (a T-lymphoblastoid line, JURKAT and a B lymphoid line, CCRF-SB) were tested under conditions of normobaric hyperoxia (FiO2 > 60%, ± 18h) and compared to a standard group (FiO2 = 21%). We tested for apoptosis using a caspase-3 assay. Cell morphology was evaluated by cytospin, microphotography after coloration, and analysis by a fractal dimension calculation software. Our results showed that exposure of cell cultures to transient normobaric hyperoxia induced apoptosis (elevated caspase-3) as well as significant and precocious modifications in cell complexity, as highlighted by increased fractal dimensions in both cell lines. These features are associated with changes in structure (pycnotic nucleus and apoptosis) recorded by microscopic analysis. Such morphological alterations could be due to several molecular mechanisms and rearrangements in the cancer cell, leading to cell cycle inhibition and apoptosis as shown by caspase-3 activity. T cells seem less resistant to hyperoxia than B cells.
Collapse
|
15
|
Qin Z, Ren G, Yuan J, Chen H, Lu Y, Li N, Zhang Y, Chen X, Zhao D. Systemic Evaluation on the Pharmacokinetics of Platinum-Based Anticancer Drugs From Animal to Cell Level: Based on Total Platinum and Intact Drugs. Front Pharmacol 2020; 10:1485. [PMID: 31969818 PMCID: PMC6960190 DOI: 10.3389/fphar.2019.01485] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 11/15/2019] [Indexed: 12/20/2022] Open
Abstract
Cisplatin, carboplatin, and oxaliplatin are the common platinum-based anticancer drugs widely used in the chemotherapeutic treatment of solid tumors in clinic. However, the comprehensive pharmacokinetics of platinum-based anticancer drugs has not been fully understood yet. This leads to many limitations for the further studies on their pharmacology and toxicology. In this study, we conduct a systemic evaluation on the pharmacokinetics of three platinum analogues at animal and cell levels, with quantification of both total platinum and intact drugs. A detailed animal study to address and compare the different pharmacokinetic behaviors of three platinum analogues has been conducted in three biological matrices: blood, plasma, and ultrafiltrate plasma. Carboplatin showed an obviously different pharmacokinetic characteristic from cisplatin and oxaliplatin. On the one hand, carboplatin has the highest proportion of Pt distribution in ultrafiltrate plasma. On the other hand, carboplatin has the highest intact drug exposure and longest intact drug elimination time in blood, plasma, and ultrafiltrate plasma, which may explain its high hematotoxicity. Additionally, the cellular and subcellular pharmacokinetics of oxaliplatin in two colon cancer HCT-116/LOVO cell lines has been elucidated for the first time. The biotransformation of intact oxaliplatin in cells was rapid with a fast elimination, however, the generated platinum-containing metabolites still exist within cells. The distribution of total platinum in the cytosol is higher than in the mitochondria, followed by the nucleus. Enrichment of platinum in mitochondria may affect the respiratory chain or energy metabolism, and further lead to cell apoptosis, which may indicate mitochondria as another potential target for efficacy and toxicity of oxaliplatin.
Collapse
Affiliation(s)
- Zhiying Qin
- Clinical Pharmacokinetics Laboratory, China Pharmaceutical University, Nanjing, China
| | - Guanghui Ren
- Clinical Pharmacokinetics Laboratory, China Pharmaceutical University, Nanjing, China
| | - Jinjie Yuan
- Clinical Pharmacokinetics Laboratory, China Pharmaceutical University, Nanjing, China
| | - Huili Chen
- School of Engineering & Applied Science, Yale University, New Haven, CT, United States
| | - Yang Lu
- Clinical Pharmacokinetics Laboratory, China Pharmaceutical University, Nanjing, China
| | - Ning Li
- National Experimental Teaching Demonstration Center of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yongjie Zhang
- Clinical Pharmacokinetics Laboratory, China Pharmaceutical University, Nanjing, China
| | - Xijing Chen
- Clinical Pharmacokinetics Laboratory, China Pharmaceutical University, Nanjing, China
| | - Di Zhao
- Clinical Pharmacokinetics Laboratory, China Pharmaceutical University, Nanjing, China
| |
Collapse
|