1
|
Tang F, Liu D, Zhang L, Xu LY, Zhang JN, Zhao XL, Ao H, Peng C. Targeting endothelial cells with golden spice curcumin: A promising therapy for cardiometabolic multimorbidity. Pharmacol Res 2023; 197:106953. [PMID: 37804925 DOI: 10.1016/j.phrs.2023.106953] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 09/20/2023] [Accepted: 10/04/2023] [Indexed: 10/09/2023]
Abstract
Cardiometabolic multimorbidity (CMM) is an increasingly significant global public health concern. It encompasses the coexistence of multiple cardiometabolic diseases, including hypertension, stroke, heart disease, atherosclerosis, and T2DM. A crucial component to the development of CMM is the disruption of endothelial homeostasis. Therefore, therapies targeting endothelial cells through multi-targeted and multi-pathway approaches hold promise for preventing and treatment of CMM. Curcumin, a widely used dietary supplement derived from the golden spice Carcuma longa, has demonstrated remarkable potential in treatment of CMM through its interaction with endothelial cells. Numerous studies have identified various molecular targets of curcumin (such as NF-κB/PI3K/AKT, MAPK/NF-κB/IL-1β, HO-1, NOs, VEGF, ICAM-1 and ROS). These findings highlight the efficacy of curcumin as a therapeutic agent against CMM through the regulation of endothelial function. It is worth noting that there is a close relationship between the progression of CMM and endothelial damage, characterized by oxidative stress, inflammation, abnormal NO bioavailability and cell adhesion. This paper provides a comprehensive review of curcumin, including its availability, pharmacokinetics, pharmaceutics, and therapeutic application in treatment of CMM, as well as the challenges and future prospects for its clinical translation. In summary, curcumin shows promise as a potential treatment option for CMM, particularly due to its ability to target endothelial cells. It represents a novel and natural lead compound that may offer significant therapeutic benefits in the management of CMM.
Collapse
Affiliation(s)
- Fei Tang
- Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Dong Liu
- Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Li Zhang
- Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Li-Yue Xu
- Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Jing-Nan Zhang
- Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Xiao-Lan Zhao
- Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Hui Ao
- Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Cheng Peng
- Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| |
Collapse
|
2
|
Ishii S, Ichimura T, Ichikura D. Prevention of vasculitis and vascular pain by side route administration of vinorelbine: A case report. Clin Case Rep 2023; 11:e7258. [PMID: 37151952 PMCID: PMC10160809 DOI: 10.1002/ccr3.7258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 04/08/2023] [Indexed: 05/09/2023] Open
Abstract
A 49-year-old female with non-small-cell lung cancer was placed on adjuvant chemotherapy with vinorelbine (25 mg/m2: Day 1.8) and cisplatin (80 mg/m2: Day 1). The simultaneous intravenous infusion of vinorelbine from the side route and 500 mL of saline from the main route successfully prevented vasculitis and vascular pain.
Collapse
Affiliation(s)
- Shunichi Ishii
- Department of Hospital PharmaceuticsSchool of Pharmacy, Showa UniversityTokyoJapan
- Department of Pharmacy ServicesShowa University Northern Yokohama HospitalKanagawaJapan
| | - Takenori Ichimura
- Department of Hospital PharmaceuticsSchool of Pharmacy, Showa UniversityTokyoJapan
- Department of Pharmacy ServicesShowa University Northern Yokohama HospitalKanagawaJapan
| | - Daisuke Ichikura
- Department of Hospital PharmaceuticsSchool of Pharmacy, Showa UniversityTokyoJapan
- Department of Pharmacy ServicesShowa University Northern Yokohama HospitalKanagawaJapan
| |
Collapse
|
3
|
The Role of PKC and HIF-1 and the Effect of Traditional Chinese Medicinal Compounds on Cerebral Ischemia-Reperfusion Injury. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:1835898. [PMID: 35265143 PMCID: PMC8898791 DOI: 10.1155/2022/1835898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 01/26/2022] [Accepted: 02/07/2022] [Indexed: 11/25/2022]
Abstract
Neuronal death occurs during cerebral ischemia. However, when hemoperfusion and oxygen supply are resumed to the ischemic focus of the brain tissue, the brain tissue damage is further aggravated, resulting in cerebral ischemia-reperfusion injury (CIRI) to the patients. Protein kinase C (PKC) plays an important role in CIRI. Through the IP3/DAG/Ca2+ signaling pathway, it promotes the influx of calcium ions in neurons and causes calcium overload, which aggravates the damage. At the same time, when brain cells are hypoxic, hypoxia-inducible factor-1 (HIF-1) is expressed, which regulates the expression of Bcl-2 and Bax through the PI3K/Akt signaling pathway and reduces nerve cell injury. It also fights hypoxic-ischemic injury by increasing the production of vascular endothelial growth factor (VEGF) to promote blood vessel formation. The PKC and HIF-1 signaling pathways are also linked to CIRI. HIF-1 activates the PKC and ERK pathways via the upregulation of VEGF, leading to increased Cx43 phosphorylation and dysfunction and aggravating CIRI. Existing studies have shown that certain traditional Chinese medicine (TCM) compounds regulate the PKC and HIF-1 signaling pathways and alleviate CIRI. These compounds downregulate the PKC and the activity of the PKC-related signaling pathways to alleviate CIRI. They can also promote the expression of HIF-1, increase the content of VEGF in ischemic tissues to promote the generation of blood vessels, and improve microcirculation. TCM compounds can inhibit the cascade of reactions underlying disease occurrence and development by targeting multiple components using different herbal formulations to improve the structural and material changes in the brain cells, which alleviate CIRI and protect the brain tissue. This study briefly describes the role of PKC and HIF-1, their relationship in CIRI, and the effect of TCM on them.
Collapse
|
4
|
Fen Y, Xinxin M, Yalan Y, Xu Z, Zhongsheng L, Shixiong S, Xinyan C, Zhenzhen W, Wei Z, Xiaolei W. The effect of a low-color-temperature-based yellow light source on the prevention of phlebitis induced by chemotherapy. Biomater Sci 2022; 10:909-914. [PMID: 35079753 DOI: 10.1039/d1bm01189h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
In recent years, light therapy has been gradually applied to the treatment of inflammation. Different from conventional high-color-temperature light sources, low-color-temperature yellow light (1900 K) without a blue light spectrum was selected as the light source to research its preventive effects on chemotherapy-induced phlebitis in this study. Based on a series of inflammatory characterization experiments, the results manifested that the reasonable utilization of 1900 K yellow light had a good effect on the prevention of phlebitis. This study shows that this is a feasible and promising method for preventing phlebitis and relieving pain, while providing a theoretical basis for the further investigation of the anti-inflammatory effects on phlebitis.
Collapse
Affiliation(s)
- Yu Fen
- College of Chemistry, Nanchang University, Nanchang, Jiangxi, 330088, P.R.China.,Institute of Advanced Materials, East China Jiaotong University, Nanchang, Jiangxi, 330013, P.R.China
| | - Miao Xinxin
- The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330088, P.R.China
| | - Yang Yalan
- Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, 330088, P.R.China
| | - Zhao Xu
- Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, 330088, P.R.China
| | - Lv Zhongsheng
- Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, 330088, P.R.China
| | - Shen Shixiong
- Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, 330088, P.R.China
| | - Cheng Xinyan
- College of Chemistry, Nanchang University, Nanchang, Jiangxi, 330088, P.R.China
| | - Weng Zhenzhen
- College of Chemistry, Nanchang University, Nanchang, Jiangxi, 330088, P.R.China
| | - Zhang Wei
- College of Chemistry, Nanchang University, Nanchang, Jiangxi, 330088, P.R.China
| | - Wang Xiaolei
- College of Chemistry, Nanchang University, Nanchang, Jiangxi, 330088, P.R.China.,Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi, 330088, P.R.China
| |
Collapse
|
5
|
He Z, Zhang Y, Zhang H, Zhou C, Ma Q, Deng P, Lu M, Mou Z, Lin M, Yang L, Li Y, Yue Y, Pi H, Lu Y, He M, Zhang L, Chen C, Zhou Z, Yu Z. NAC antagonizes arsenic-induced neurotoxicity through TMEM179 by inhibiting oxidative stress in Oli-neu cells. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 223:112554. [PMID: 34332247 DOI: 10.1016/j.ecoenv.2021.112554] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 07/20/2021] [Accepted: 07/21/2021] [Indexed: 06/13/2023]
Abstract
Arsenic is one of the most common environmental pollutants. Neurotoxicity induced by arsenic has become a major public health concern. However, the effects of arsenic-induced neurotoxicity in the brain and the underlying molecular mechanisms are not well understood. N-acetyl-cysteine (NAC) is a thiol-based antioxidant that can antagonize heavy metal-induced neurotoxicity by scavenging reactive oxygen species (ROS). Here, we used the mouse oligodendrocyte precursor cell (OPC) line Oli-neu to explore the neurotoxic effects of arsenic and the protective effects of NAC. We found that arsenic exposure decreased cell viability, increased oxidative stress, caused mitochondrial dysfunction, and led to apoptosis of Oli-neu cells. Furthermore, we revealed that NAC treatment reversed these neurotoxic effects of arsenic. TMEM179, a key membrane protein, was found highly expressed in OPCs and to be an important factor in maintaining mitochondrial functions. We found that TMEM179 played a critical role in mediating the neurotoxic effects of arsenic and the protective role of NAC. PKCβ is a downstream factor through which TMEM179 regulates the expression of apoptosis-related proteins. This study improves our understanding of the neurotoxic effects and mechanisms of arsenic exposure and the protective effects of NAC. It also identifies a potential molecular target, TMEM179, for the treatment of arsenic-induced neurotoxicity.
Collapse
Affiliation(s)
- Zhixin He
- Department of Occupational Health, Army Medical University, 400038, Chongqing, People's Republic of China
| | - Yajing Zhang
- School of Medicine, Guangxi University, 530004, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Huijie Zhang
- School of Medicine, Guangxi University, 530004, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Chao Zhou
- Department of Occupational Health, Army Medical University, 400038, Chongqing, People's Republic of China
| | - Qinlong Ma
- Department of Occupational Health, Army Medical University, 400038, Chongqing, People's Republic of China
| | - Ping Deng
- Department of Occupational Health, Army Medical University, 400038, Chongqing, People's Republic of China
| | - Muxue Lu
- School of Medicine, Guangxi University, 530004, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Zhenlin Mou
- School of Medicine, Guangxi University, 530004, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Min Lin
- Department of Occupational Health, Army Medical University, 400038, Chongqing, People's Republic of China
| | - Lingling Yang
- Department of Occupational Health, Army Medical University, 400038, Chongqing, People's Republic of China
| | - Yanqi Li
- Department of Occupational Health, Army Medical University, 400038, Chongqing, People's Republic of China
| | - Yang Yue
- Department of Occupational Health, Army Medical University, 400038, Chongqing, People's Republic of China
| | - Huifeng Pi
- Department of Occupational Health, Army Medical University, 400038, Chongqing, People's Republic of China
| | - Yonghui Lu
- Department of Occupational Health, Army Medical University, 400038, Chongqing, People's Republic of China
| | - Mindi He
- Department of Occupational Health, Army Medical University, 400038, Chongqing, People's Republic of China
| | - Lei Zhang
- Department of Occupational Health, Army Medical University, 400038, Chongqing, People's Republic of China
| | - Chunhai Chen
- Department of Occupational Health, Army Medical University, 400038, Chongqing, People's Republic of China.
| | - Zhou Zhou
- Department of Environmental Medicine, School of Public Health, and Department of Emergency Medicine, First Affiliated Hospital, School of Medicine, Zhejiang University, 310058 Hangzhou, People's Republic of China.
| | - Zhengping Yu
- Department of Occupational Health, Army Medical University, 400038, Chongqing, People's Republic of China.
| |
Collapse
|
6
|
Yu L, Zhang Y, Zhao X, Wan H, He Y, Jin W. Guhong Injection Alleviates Cerebral Ischemia-Reperfusion Injury via the PKC/HIF-1α Pathway in Rats. Front Pharmacol 2021; 12:716121. [PMID: 34539402 PMCID: PMC8443782 DOI: 10.3389/fphar.2021.716121] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 08/19/2021] [Indexed: 01/01/2023] Open
Abstract
Guhong injection (GHI) is a drug for ischemic stroke created by combining safflower, a traditional Chinese medicine, and aceglutamide, a Western medicine. In this study, we investigated the curative effect of GHI on cerebral ischemia–reperfusion (I/R) injury via the PKC/HIF-1α pathway in rats. Adult male Sprague Dawley rats were randomly divided into seven groups: sham-operated, middle cerebral artery occlusion (MCAO), GHI, nimodipine injection (NMDP), MCAO + LY317615 (PKC inhibitor), GHI + LY317615, and NMDP + LY317615. After establishing an MCAO rat model, we performed neurological deficit testing, 2,3,5-triphenyltetrazolium chloride staining, hematoxylin and eosin (HE) staining, enzyme-linked immunosorbent assay, Western blotting, and q-PCR to detect the brain damage in rats. Compared with the MCAO group, the GHI and GHI + LY317615 group showed neurological damage amelioration as well as decreases in serum hypoxia-inducible factor-1α (HIF-1α), protein kinase C (PKC), and erythropoietin levels; brain HIF-1α and inducible nitric oxide synthase protein expression; and brain HIF-1α and NOX-4 mRNA expression. These effects were similar to those in the positive control groups NMDP and NMDP + LY317615. Thus, our results confirmed GHI can ameliorate cerebral I/R injury in MCAO rats possibly via the PKC/HIF-1α pathway.
Collapse
Affiliation(s)
- Li Yu
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yangyang Zhang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xixi Zhao
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Haitong Wan
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yu He
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Weifeng Jin
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
7
|
Ozawa N, Hase T, Hatta T, Sagara A, Ichikawa K, Miyazaki M, Yogo N, Ando M, Hashimoto N, Yamada K, Hasegawa Y. Comparing incidences of infusion site reactions between brand-name and generic vinorelbine in patients with non-small cell lung cancer. Br J Clin Pharmacol 2020; 87:1318-1326. [PMID: 32790917 DOI: 10.1111/bcp.14516] [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: 01/20/2020] [Revised: 07/05/2020] [Accepted: 08/06/2020] [Indexed: 11/26/2022] Open
Abstract
AIM This study aimed to compare the incidence of infusion site reactions (ISRs) induced by intravenous administration of brand-name and generic vinorelbine (VNR) for treating non-small cell lung cancer. METHOD This single-centre retrospective cohort study was conducted by medical chart review of VNR infusions. ISRs were defined as symptoms around the infusion site, including pain, redness and swelling. ISRs requiring treatment were defined as ISRs requiring treatments including steroid ointments, vein repuncture and local steroid injections. RESULTS In all, 1973 VNR infusions were administered to 340 patients (brand-name 141 patients, generic 199 patients). ISRs and ISRs requiring treatment were observed in 161 and 100 patients, respectively. The ISR incidence per patient and per injection was significantly higher in generic VNR-treated patients than in brand-name VNR-treated patients (53.3% vs 39.0%, P = 0.0112 and 15.0% vs 9.9%, P = 0.0008, respectively). The frequency of ISRs requiring treatment was also significantly higher in the generic group (per patient 36.7% vs 19.2%, P = 0.0005; per injection 11.3% vs 5.5%, P < 0.0001). Multivariate analysis revealed that generic VNR was significantly associated with an increased risk of ISRs (per patient adjusted odds ratio [AOR] 1.775, P = 0.0155; per injection AOR 1.672, P = 0.004) and ISRs requiring treatment (per patient AOR 2.422, P = 0.0012; per injection AOR 2.286, P = 0.001). CONCLUSION Intravenous infusion of generic VNR was associated with an increased risk of ISRs. Further research is needed to elucidate the mechanism underlying the increased incidence of ISRs with generic VNR.
Collapse
Affiliation(s)
- Naoya Ozawa
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tetsunari Hase
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takahiro Hatta
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Atsunobu Sagara
- Department of Pharmacy, Nagoya University Hospital, Nagoya, Japan
| | - Kazuya Ichikawa
- Department of Pharmacy, Nagoya University Hospital, Nagoya, Japan
| | | | - Naoyuki Yogo
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masahiko Ando
- Centre for Advanced Medicine and Clinical Research, Nagoya University Hospital, Nagoya, Japan
| | - Naozumi Hashimoto
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kiyofumi Yamada
- Department of Pharmacy, Nagoya University Hospital, Nagoya, Japan
| | - Yoshinori Hasegawa
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan.,National Hospital Organization, Nagoya Medical Center, Nagoya, Japan
| |
Collapse
|
8
|
Mendez N, Alarcón P, Millán C, Burgos RA, Morera FJ, Ojeda J. Vincristine, carboplatin and cisplatin increase oxidative burst induced by PAF in canine neutrophils. Vet Immunol Immunopathol 2020; 221:110011. [PMID: 32007672 DOI: 10.1016/j.vetimm.2020.110011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 12/12/2019] [Accepted: 01/14/2020] [Indexed: 01/05/2023]
Abstract
Myelosupression resulting from chemotherapy has been widely described in veterinary medicine; however, there is limited information relating to alterations in neutrophil function after chemotherapy in dogs with cancer. The aim of this study was to determine the non-proliferative effects of vincristine, carboplatin, and cisplatin on canine neutrophils by evaluating activation of oxidative and non-oxidative responses. Neutrophils were isolated from venous blood. Levels of reactive oxygen species (ROS) and metalloproteinase 9 (MMP-9) were measured in vitro during neutrophil exposure to these chemotherapeutic agents for 15 min followed by stimulation with platelet activating factor (PAF). ROS production was detected via luminescence, and MMP- 9 liberation was determined by zymography. The chemotherapeutic agents caused an increase in PAF-induced ROS production, but no change in the non-oxidative response was observed. These results suggest that these chemotherapeutic agents may act as priming agents by increasing the oxidative response. These effects could be beneficial for dogs with cancer by supporting their immune systems; however, excessive ROS liberation has been associated with inflammation, neutrophil-mediated cell injury, carcinogenesis, and metastasis. Clinical studies are necessary to evaluate the significance of these findings.
Collapse
Affiliation(s)
- Nathalia Mendez
- Veterinary Clinical Sciences, Faculty of Veterinary Sciences, Universidad Austral de Chile, Valdivia, Chile
| | - Pablo Alarcón
- Laboratory of Molecular Pharmacology, Institute of Pharmacology and Morphophysiology, Faculty of Veterinary Science, Universidad Austral de Chile, Valdivia, Chile
| | - Catalina Millán
- Applied Biochemistry Laboratory, Institute of Pharmacology and Morphophysiology, Faculty of Veterinary Science, Universidad Austral de Chile, Valdivia, Chile; School of Medicine, Medicine Faculty, Universidad Austral de Chile, Valdivia, Chile
| | - Rafael A Burgos
- Laboratory of Molecular Pharmacology, Institute of Pharmacology and Morphophysiology, Faculty of Veterinary Science, Universidad Austral de Chile, Valdivia, Chile
| | - Francisco J Morera
- Applied Biochemistry Laboratory, Institute of Pharmacology and Morphophysiology, Faculty of Veterinary Science, Universidad Austral de Chile, Valdivia, Chile
| | - Javier Ojeda
- Veterinary Clinical Sciences, Faculty of Veterinary Sciences, Universidad Austral de Chile, Valdivia, Chile.
| |
Collapse
|
9
|
Morimoto Y, Miyawaki K, Seki R, Watanabe K, Hirohara M, Shinohara T. Risk factors for venous irritation in patients receiving vinorelbine: a retrospective study. J Pharm Health Care Sci 2018; 4:26. [PMID: 30288295 PMCID: PMC6166274 DOI: 10.1186/s40780-018-0122-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 08/28/2018] [Indexed: 05/29/2023] Open
Abstract
Background Vinorelbine is known to be effective in the treatment of non-small cell lung cancer and breast cancer. However, venous irritation is a common side effect. Although there have been some reports on risk factors for venous irritation in patients receiving vinorelbine, the factors evaluated have been limited and the results inconclusive. The aim of this study was to identify risk factors for venous irritation in patients receiving vinorelbine, and factors likely associated with venous irritation, including new factors such as hot compress with a hot towel for prevention of venous irritation. Methods We retrospectively reviewed patients treated with vinorelbine at Kyorin University Hospital, Japan, between March 2013 and December 2016 and divided them into the two groups according to whether or not they had venous irritation. Clinical characteristics were compared between the two groups. Results Venous irritation occurred in 24 (38.1%) of 63 patients who received vinorelbine. The median number of times vinorelbine was administered before onset of venous irritation was 3 (range 1–14). The group with venous irritation had a significantly lower body surface area than the group without venous irritation (p = 0.035). Low body surface area was also the only significant risk factor for vinorelbine-associated venous irritation in multivariate analysis (adjusted odds ratio 70.42 per 1 m2decrement, 95% confidence interval 1.54–3236.25, p = 0.029). There was no association between the occurrence of venous irritation and the other covariates, such as use of a hot compress, history of diabetes mellitus, or use of a generic formulation of vinorelbine. Conclusion Low body surface area may be a risk factor for venous irritation in patients receiving vinorelbine. Use of hot compress with a hot towel did not prevent venous irritation.
Collapse
Affiliation(s)
- Yoshihito Morimoto
- 1Education and Research Center for Clinical Pharmacy, Showa Pharmaceutical University, 3-3165 Higashi-Tamagawagakuen, Machida, Tokyo, 194-8543 Japan
| | - Kumika Miyawaki
- 2Department of Pharmacy, Kyorin University Hospital, 6-20-2 Shinkawa, Mitaka, Tokyo, 181-8611 Japan
| | - Reisuke Seki
- 2Department of Pharmacy, Kyorin University Hospital, 6-20-2 Shinkawa, Mitaka, Tokyo, 181-8611 Japan
| | - Kazuhiro Watanabe
- 1Education and Research Center for Clinical Pharmacy, Showa Pharmaceutical University, 3-3165 Higashi-Tamagawagakuen, Machida, Tokyo, 194-8543 Japan
| | - Masayoshi Hirohara
- 1Education and Research Center for Clinical Pharmacy, Showa Pharmaceutical University, 3-3165 Higashi-Tamagawagakuen, Machida, Tokyo, 194-8543 Japan
| | - Takao Shinohara
- 2Department of Pharmacy, Kyorin University Hospital, 6-20-2 Shinkawa, Mitaka, Tokyo, 181-8611 Japan
| |
Collapse
|
10
|
Research Progress on Signaling Pathway-Associated Oxidative Stress in Endothelial Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:7156941. [PMID: 28503253 PMCID: PMC5414589 DOI: 10.1155/2017/7156941] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 02/01/2017] [Accepted: 02/16/2017] [Indexed: 12/31/2022]
Abstract
Studying the mechanisms of oxidative stress in endothelial cells is vital to the discovery of novel drugs for the treatment of cardiovascular disease. This article reviews the progress within the field of the role of oxidative responses in the physiology and growth of endothelial cells and emphasizes the effects of several main signal pathways involved in the oxidative stress of endothelial cells. Herein, we aim to provide scientific direction that can serve as a basis for researchers specializing in the signaling pathway of oxidative stress.
Collapse
|
11
|
Protein kinase C phosphorylates AMP-activated protein kinase α1 Ser487. Biochem J 2016; 473:4681-4697. [PMID: 27784766 PMCID: PMC5147050 DOI: 10.1042/bcj20160211] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Revised: 10/23/2016] [Accepted: 10/25/2016] [Indexed: 12/16/2022]
Abstract
The key metabolic regulator, AMP-activated protein kinase (AMPK), is reported to be down-regulated in metabolic disorders, but the mechanisms are poorly characterised. Recent studies have identified phosphorylation of the AMPKα1/α2 catalytic subunit isoforms at Ser487/491, respectively, as an inhibitory regulation mechanism. Vascular endothelial growth factor (VEGF) stimulates AMPK and protein kinase B (Akt) in cultured human endothelial cells. As Akt has been demonstrated to be an AMPKα1 Ser487 kinase, the effect of VEGF on inhibitory AMPK phosphorylation in cultured primary human endothelial cells was examined. Stimulation of endothelial cells with VEGF rapidly increased AMPKα1 Ser487 phosphorylation in an Akt-independent manner, without altering AMPKα2 Ser491 phosphorylation. In contrast, VEGF-stimulated AMPKα1 Ser487 phosphorylation was sensitive to inhibitors of protein kinase C (PKC) and PKC activation using phorbol esters or overexpression of PKC-stimulated AMPKα1 Ser487 phosphorylation. Purified PKC and Akt both phosphorylated AMPKα1 Ser487 in vitro with similar efficiency. PKC activation was associated with reduced AMPK activity, as inhibition of PKC increased AMPK activity and phorbol esters inhibited AMPK, an effect lost in cells expressing mutant AMPKα1 Ser487Ala. Consistent with a pathophysiological role for this modification, AMPKα1 Ser487 phosphorylation was inversely correlated with insulin sensitivity in human muscle. These data indicate a novel regulatory role of PKC to inhibit AMPKα1 in human cells. As PKC activation is associated with insulin resistance and obesity, PKC may underlie the reduced AMPK activity reported in response to overnutrition in insulin-resistant metabolic and vascular tissues.
Collapse
|
12
|
Mhaidat NM, Alzoubi KH, Khabour OF, Alawneh KZ, Raffee LA, Alsatari ES, Hussein EI, Bani-Hani KE. Assessment of genotoxicity of vincristine, vinblastine and vinorelbine in human cultured lymphocytes: a comparative study. Balkan J Med Genet 2016; 19:13-20. [PMID: 27785403 PMCID: PMC5026275 DOI: 10.1515/bjmg-2016-0002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Vincristine (VCR), vinblastine (VBL) and vinorelbine (VRL) are anticancer agents from the Vinca alkaloid family that have the potential to induce genotoxic effect. The aim of the present study was to compare the genotoxic effect of VCR, VBL and VRL. Levels of 8-hydroxy-2-deoxy guanosine (8-OHdG) and sister chromatid exchanges (SCEs) were measured in cultured human blood lymphocytes treated with VCR, VBL and VRL at concentrations of 0.01 and 0.1 μg/mL. Results showed that VCR, VBL and VRL significantly increased the 8-OHdG levels (p <0.05), whereas it did not cause a significant increase in the frequencies of SCEs in human blood lymphocytes as compared to controls. On the other hand, all three agents significantly increased cells mitotic index (p <0.05). At both tested concentrations, the magnitude of the increase in 8-OHdG was VBL>VCR>VRL. In conclusion, VCR, VBL and VRL induce DNA damage as indicated by the increase in the 8-OHdG biomarker but with different magnitude.
Collapse
Affiliation(s)
- N M Mhaidat
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - K H Alzoubi
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - O F Khabour
- Department of Medical Laboratory Sciences, Faculty of Applied Medical sciences, Jordan University of Science and Technology, Irbid, Jordan
| | - K Z Alawneh
- Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - L A Raffee
- Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - E S Alsatari
- Department of Applied Biological Sciences, Faculty of Science and Arts, Jordan University of Science and Technology, Irbid, Jordan
| | - E I Hussein
- Department of Biology, Faculty of Science, Yarmouk University, Irbid, Jordan
| | - K E Bani-Hani
- Faculty of Medicine, Hashemite University, Zarqaa, Jordan
| |
Collapse
|
13
|
Biziota E, Briasoulis E, Mavroeidis L, Marselos M, Harris AL, Pappas P. Cellular and molecular effects of metronomic vinorelbine and 4-O-deacetylvinorelbine on human umbilical vein endothelial cells. Anticancer Drugs 2016; 27:216-24. [PMID: 26629767 PMCID: PMC4733659 DOI: 10.1097/cad.0000000000000319] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 10/25/2015] [Indexed: 12/31/2022]
Abstract
Metronomic oral vinorelbine (VRL; Navelbine) was shown in clinical trials to yield sustainable antitumor activity possibly through antiangiogenic mechanisms. We investigated the effects of protracted low-dose VRL on human umbilical vein endothelial cells, compared with a conventional chemotherapy model. Human umbilical vein endothelial cell cultures were treated with different concentrations of VRL (0.001 nmol/l to 1 mmol/l) for 4, 24 and 96 h. The effects of different drug concentrations on cell growth, cell cycle, apoptosis and expression of the angiogenesis-modulating genes interleukin-8, cyclooxygenase-2, CD36 and peroxisome proliferator-activated receptor γ were assessed using the metronomic or conventional chemotherapy model. Apoptosis and cell-cycle effects were assessed by flow cytometry. Gene expression was measured at the transcript level by quantitative reverse transcriptase-PCR, protein expression by immunoblotting and levels of proteins secreted in the cell medium by enzyme-linked immunosorbent assay. Activation of the nuclear factor-κB pathway was investigated by immunoblot analysis of cytosolic and nuclear protein extracts. The half-maximal inhibitory concentrations (IC50) of VRL at 96 h were four orders lower compared with those after a 24-h exposure (1.23 nmol/l vs. 32 mmol/l for VRL). Drug concentrations at high nanomolar levels and above, which are relevant to conventional pulsatile dosing of VRL, induced a dose-dependent and nuclear factor-κB-related increase in proangiogenic interleukin-8 and cyclooxygenase-2 and a decrease in the thrombospondin-1 receptor CD36 and peroxisome proliferator-activated receptor γ at mRNA and protein levels. In contrast, the opposite was evident with protracted picomolar to low nanomolar concentrations (metronomic dosing). Our data provide experimental support for metronomic VRL by showing that a protracted low dose outperforms pulsed high-dose administration in inducing antiangiogenic effects in proliferating human endothelial cells.
Collapse
Affiliation(s)
| | - Evangelos Briasoulis
- Interscience Molecular Laboratory, Cancer Biobank Center
- Department of Haematology, Division of Medicine, School of Life Sciences, Ioannina, Greece
| | - Leonidas Mavroeidis
- Department of Pharmacology, School of Medicine
- Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | | | - Adrian L. Harris
- Molecular Oncology Laboratories, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | | |
Collapse
|
14
|
Liver Trapping of 99mTc Macroaggregated Albumin During Ventilation/Perfusion Scintigraphy in a Patient With Superior Vena Cava Stenosis as Demonstrated by SPECT/CT. Clin Nucl Med 2015; 40:e366-9. [DOI: 10.1097/rlu.0000000000000791] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
15
|
Homocysteine facilitates LOX-1 activation and endothelial death through the PKCβ and SIRT1/HSF1 mechanism: relevance to human hyperhomocysteinaemia. Clin Sci (Lond) 2015; 129:477-87. [DOI: 10.1042/cs20150127] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 05/15/2015] [Indexed: 12/20/2022]
Abstract
Our study demonstrates a new direction for LOX-1 regulation by modulating the PKCβ/NADPH oxidase/SIRT1/HSF1 pathway, which affects hyperhomocysteinaemia-induced endothelial cell dysfunction and apoptosis.
Collapse
|
16
|
Yamada T. [Pharmacological study and pharmaceutical intervention to reduce intravenous injection-induced vascular injury]. YAKUGAKU ZASSHI 2015; 135:465-72. [PMID: 25759054 DOI: 10.1248/yakushi.14-00161] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Intravenous injection often causes vascular injury such as venous irritation, vascular pain, and phlebitis. Vascular injury deteriorates the patient's QOL and sometimes limits the continuation of injection therapy. Pharmaceutical intervention and pharmacological mechanisms used to reduce vascular injury induced by vinorelbine and epirubicin were reviewed. A multivariate logistic regression analysis revealed that the dose of vinorelbine (≥40 mg) was a significant predictor for venous irritation. Alteration of the volume of normal saline for vinorelbine dissolution, from 50 to 100 mL, significantly decreased the grade of venous irritation. On the other hand, the phlebitis scores were significantly higher in patients treated with epirubicin ready-to-use solution compared with lyophilized powder. The change of formulation of epirubicin to lyophilized powder decreased the risk of venous irritation. The concentration inducing 50% cell viability inhibition was lower in the order of vesicant, irritant, and nonvesicant drugs on porcine aorta endothelial cells (PAECs), suggesting that the injuring effects of anticancer drugs on PAECs may be relevant as an indicator of the frequency of their vascular injury. The exposure to vinorelbine of PAECs rapidly depleted intracellular glutathione levels and increased intracellular reactive oxygen species production. Moreover, exposure to epirubicin increased intracellular lipid peroxide levels and enhanced the phosphorylation of p38 mitogen-activated protein kinase. These results demonstrate that oxidative stress plays an important role in vinorelbine- and epirubicin-induced endothelial cell injury, and may therefore increase the potential for vascular injury upon intravenous injection.
Collapse
|
17
|
Hung CH, Chan SH, Chu PM, Tsai KL. Docetaxel Facilitates Endothelial Dysfunction through Oxidative Stress via Modulation of Protein Kinase C Beta: The Protective Effects of Sotrastaurin. Toxicol Sci 2015; 145:59-67. [PMID: 25634538 DOI: 10.1093/toxsci/kfv017] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Docetaxel (DTX), a taxane drug, has widely been used as an anticancer or antiangiogenesis drug. However, DTX caused side effects, such as vessel damage and phlebitis, which may reduce its clinical therapeutic efficacy. The molecular mechanisms of DTX that cause endothelial dysfunction remain unclear. The aim of this study as to validate the probable mechanisms of DTX-induced endothelial dysfunction in endothelial cells. Human umbilical vein endothelial cells (HUVECs) were stimulated with DTX (2.5, 5, and 10nM) for 24 h to induce endothelial dysfunction. Stimulation with DTX reduced cell viability in a concentration- and time-dependent manner. DTX upregulated caspase-3 activity and TUNEL-positive cells. DTX treatment also increased PKCβ phosphorylation levels and NADPH oxidase activity, which resulted in ROS formation. However, all of these findings were reversed by PKCβ inhibition and NADPH oxidase repression. Finally, we demonstrated that sotrastaurin (AEB-071), a new PKCβ inhibitor, mitigated DTX-induced oxidative injury in endothelial cells. Our findings from this study provide a probable molecular mechanism of DTX-induced oxidative injury in endothelial cells and a new clinical and therapeutic approach for preventing DTX-mediated vessel injury.
Collapse
Affiliation(s)
- Ching-Hsia Hung
- *Department of Physical Therapy, National Cheng Kung University, Tainan, Taiwan, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan and Department of Anatomy, School of Medicine, China Medical University, Taichung, Taiwan
| | - Shih-Hung Chan
- *Department of Physical Therapy, National Cheng Kung University, Tainan, Taiwan, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan and Department of Anatomy, School of Medicine, China Medical University, Taichung, Taiwan
| | - Pei-Ming Chu
- *Department of Physical Therapy, National Cheng Kung University, Tainan, Taiwan, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan and Department of Anatomy, School of Medicine, China Medical University, Taichung, Taiwan
| | - Kun-Ling Tsai
- *Department of Physical Therapy, National Cheng Kung University, Tainan, Taiwan, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan and Department of Anatomy, School of Medicine, China Medical University, Taichung, Taiwan
| |
Collapse
|
18
|
Tsai KL, Huang PH, Kao CL, Leu HB, Cheng YH, Liao YW, Yang YP, Chien Y, Wang CY, Hsiao CY, Chiou SH, Chen JW, Lin SJ. Aspirin attenuates vinorelbine-induced endothelial inflammation via modulating SIRT1/AMPK axis. Biochem Pharmacol 2013; 88:189-200. [PMID: 24345330 DOI: 10.1016/j.bcp.2013.12.005] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2013] [Revised: 11/24/2013] [Accepted: 12/03/2013] [Indexed: 12/20/2022]
Abstract
Vinorelbine (VNR), a semisynthetic vinca alkaloid acquired from vinblastine, is frequently used as the candidate for intervention of solid tumors. Nevertheless, VNR-caused endothelial injuries may lead a mitigative effect of clinical treatment efficiency. A growing body of evidence reveals that aspirin is a potent antioxidant and anti-inflammation drug. We investigated whether aspirin attenuate VNR-induced endothelial dysfunction. Human endothelial cells (EA.hy 926) were treated with VNR to cause endothelial inflammation. Western blotting, ROS assay, ELISA were used to confirm the anti-inflammatory effect of aspirin. We confirmed that VNR suppresses SIRT1 expression, reduced LKB1 and AMPK phosphorylation as well as enriched PKC activation in treated endothelial cells. Furthermore, the membrane translocation assay displayed that the levels of NADPH oxidase subunits p47phox and Rac-1 in membrane fractions of endothelial cells were higher in cells that had been treated with VNR for than in untreated cells. We corroborated that treatment of Aspirin significantly diminishes VNR-repressed SIRT1, LKB1 and AMPK phosphorylation and VNR-promoted NADPH oxidase activation, however, those findings were vanished by SIRT1 and AMPK siRNAs. Our data also shown that Aspirin represses VNR-activated TGF-beta-activated kinase-1 (TAK1) activation, inhibited the interaction of TAK1/TAK-binding protein1 (TAB1), suppressed NF-kappa B activation and pro-inflammatory cytokine secretion. We demonstrated a novel connection between VNR-caused oxidative damages and endothelial dysfunction, and provide further insight into the protective effects of aspirin in VNR-caused endothelial dysfunction.
Collapse
Affiliation(s)
- Kun-Ling Tsai
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Po-Hsun Huang
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan; Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chung-Lan Kao
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan; Department of Physical Medicine and Rehabilitation, Taipei, Taiwan
| | - Hsin-Bang Leu
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan; Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yung-Hsin Cheng
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Pharmacology, National Yang-Ming University, Taipei, Taiwan
| | - Yi-Wen Liao
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Pharmacology, National Yang-Ming University, Taipei, Taiwan; Department of Medical Research and Education, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yi-Ping Yang
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Pharmacology, National Yang-Ming University, Taipei, Taiwan; Department of Medical Research and Education, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yueh Chien
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Pharmacology, National Yang-Ming University, Taipei, Taiwan; Department of Medical Research and Education, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chien-Ying Wang
- Department of Emergency Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Chen-Yuan Hsiao
- Department of Surgery, National Yang-Ming University Hospital, Taipei, Taiwan
| | - Shih-Hwa Chiou
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan; Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Pharmacology, National Yang-Ming University, Taipei, Taiwan; Department of Medical Research and Education, Taipei Veterans General Hospital, Taipei, Taiwan.
| | - Jaw-Wen Chen
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Pharmacology, National Yang-Ming University, Taipei, Taiwan; Department of Medical Research and Education, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Shing-Jong Lin
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan; Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Department of Medical Research and Education, Taipei Veterans General Hospital, Taipei, Taiwan.
| |
Collapse
|
19
|
Liu YR, Huang SY, Chen JY, Wang LHC. Microtubule depolymerization activates the Epstein–Barr virus lytic cycle through protein kinase C pathways in nasopharyngeal carcinoma cells. J Gen Virol 2013; 94:2750-2758. [DOI: 10.1099/vir.0.058040-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Elevated levels of antibodies against Epstein–Barr virus (EBV) and the presence of viral DNA in plasma are reliable biomarkers for the diagnosis of nasopharyngeal carcinoma (NPC) in high-prevalence areas, such as South-East Asia. The presence of these viral markers in the circulation suggests that a minimal level of virus reactivation may have occurred in an infected individual, although the underlying mechanism of reactivation remains to be elucidated. Here, we showed that treatment with nocodazole, which provokes the depolymerization of microtubules, induces the expression of two EBV lytic cycle proteins, Zta and EA-D, in EBV-positive NPC cells. This effect was independent of mitotic arrest, as viral reactivation was not abolished in cells synchronized at interphase. Notably, the induction of Zta by nocodazole was mediated by transcriptional upregulation via protein kinase C (PKC). Pre-treatment with inhibitors for PKC or its downstream signalling partners p38 mitogen-activated protein kinase (MAPK) and c-Jun N-terminal kinase (JNK) abolished the nocodazole-mediated induction of Zta and EA-D. Interestingly, the effect of nocodazole, as well as colchicine and vinblastine, on lytic gene expression occurred only in NPC epithelial cells but not in cells derived from lymphocytes. These results establish a novel role of microtubule integrity in controlling the EBV life cycle through PKC and its downstream pathways, which represents a tissue-specific mechanism for controlling the life-cycle switch of EBV.
Collapse
Affiliation(s)
- Yi-Ru Liu
- Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu 300, Taiwan
- National Institute of Cancer Research, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli County 35053, Taiwan
- Graduate Program of Biotechnology in Medicine of National Tsing Hua University and National Health Research Institutes, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Sheng-Yen Huang
- National Institute of Cancer Research, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli County 35053, Taiwan
- Graduate Program of Biotechnology in Medicine of National Tsing Hua University and National Health Research Institutes, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Jen-Yang Chen
- National Institute of Cancer Research, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli County 35053, Taiwan
| | - Lily Hui-Ching Wang
- Department of Medical Science, National Tsing Hua University, Hsinchu 300, Taiwan
- Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu 300, Taiwan
| |
Collapse
|
20
|
Zhang J, Tong N, Chen Y, Li P, Yang S, Zhao X. Resveratrol protects against vinorelbine-induced vascular endothelial cell injury. Toxicol Mech Methods 2013; 23:665-71. [DOI: 10.3109/15376516.2013.837130] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
21
|
Wang Y, Ding M, Chaudhari S, Ding Y, Yuan J, Stankowska D, He S, Krishnamoorthy R, Cunningham JT, Ma R. Nuclear factor κB mediates suppression of canonical transient receptor potential 6 expression by reactive oxygen species and protein kinase C in kidney cells. J Biol Chem 2013; 288:12852-65. [PMID: 23525112 PMCID: PMC3642329 DOI: 10.1074/jbc.m112.410357] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Revised: 03/21/2013] [Indexed: 02/05/2023] Open
Abstract
This study was carried out to explore the molecular mechanism for down-regulation of TRPC6 expression in the reactive oxygen species (ROS)/PKC signaling in kidney cells. In cultured human mesangial cells, H2O2 and TNF-α inhibited TRPC6 mRNA expression in a time-dependent manner. Inhibition of NF-κB reversed both H2O2- and phorbol 12-myristate 13-acetate (PMA)-induced decrease in TRPC6 protein expression. Activation of NF-κB by knocking down IκBα using siRNA could mimic the suppressive effect of ROS/PKC on TRPC6. a Ca(2+) imaging study showed that activation and inhibition of NF-κB significantly decreased and increased the TRPC6-mediated Ca(2+) entry, respectively. Further experiments showed that PMA, but not its inactive analog 4α-phorbol 12, 13-didecanoate (4α-PDD), caused phosphorylation of IκBα and stimulated the nuclear translocation of NF-κB p50 and p65 subunits. The PMA-dependent IκBα phosphorylation was significantly inhibited by Gö6976. Electrophoretic mobility shift assay revealed that PMA stimulated DNA binding activity of NF-κB. Furthermore, specific knockdown of p65, but not p50, prevented an H2O2 inhibitory effect on TRPC6 protein expression, suggesting p65 as a predominant NF-κB subunit repressing TRPC6. In agreement with a major role of p65, chromatin immunoprecipitation assays showed that PMA treatment induced p65 binding to the TRPC6 promoter. Moreover, PMA treatment increased the association of p65 with histone deacetylase (HDAC) and decreased histone acetylation at the TRPC6 promoter. Consistently, knockdown of HDAC2 by siRNA or inhibition of HDAC with trichostatin A prevented a H2O2-induced decrease in TRPC6 mRNA and protein expressions, respectively. Taken together, our findings imply an important role of NF-κB in a negative regulation of TRPC6 expression at the gene transcription level in kidney cells.
Collapse
Affiliation(s)
- Yanxia Wang
- From the Department of Integrative Physiology and Cardiovascular Research Institute and
| | - Min Ding
- From the Department of Integrative Physiology and Cardiovascular Research Institute and
| | - Sarika Chaudhari
- From the Department of Integrative Physiology and Cardiovascular Research Institute and
| | - Yanfeng Ding
- From the Department of Integrative Physiology and Cardiovascular Research Institute and
| | - Joseph Yuan
- From the Department of Integrative Physiology and Cardiovascular Research Institute and
| | - Dorota Stankowska
- the Department of Cell Biology, University of North Texas Health Science Center, Fort Worth, Texas 76107
| | - Shaoqing He
- the Department of Cell Biology, University of North Texas Health Science Center, Fort Worth, Texas 76107
| | - Raghu Krishnamoorthy
- the Department of Cell Biology, University of North Texas Health Science Center, Fort Worth, Texas 76107
| | - Joseph T. Cunningham
- From the Department of Integrative Physiology and Cardiovascular Research Institute and
| | - Rong Ma
- From the Department of Integrative Physiology and Cardiovascular Research Institute and
| |
Collapse
|
22
|
Zhang Y, Cardell LO, Edvinsson L, Xu CB. MAPK/NF-κB-dependent upregulation of kinin receptors mediates airway hyperreactivity: a new perspective for the treatment. Pharmacol Res 2013; 71:9-18. [PMID: 23428345 DOI: 10.1016/j.phrs.2013.02.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Revised: 02/02/2013] [Accepted: 02/11/2013] [Indexed: 12/30/2022]
Abstract
Airway hyperreactivity (AHR) is a major feature of asthmatic and inflammatory airways. Cigarette smoke exposure, and bacterial and viral infections are well-known environmental risk factors for AHR, but knowledge about the underlying molecular mechanisms on how these risk factors lead to the development of AHR is limited. Activation of intracellular mitogen-activated protein kinase (MAPK)/nuclear factor-kappa B (NF-κB) and their related signal pathways including protein kinase C (PKC), phosphoinositide 3-kinase (PI3K) and protein kinase A (PKA) signaling pathways may result in airway kinin receptor upregulation, which is suggested to play an important role in the development of AHR. Environmental risk factors trigger the production of pro-inflammatory mediators such as tumor necrosis factor-α (TNF-α) and interleukins (ILs) that activate intracellular MAPK- and NF-κB-dependent inflammatory pathways, which subsequently lead to AHR via kinin receptor upregulation. Blockage of intracellular MAPK/NF-κB signaling prevents kinin B₁ and B₂ receptor expression in the airways, resulting in a decrease in the response to bradykinin (kinin B₂ receptor agonist) and des-Arg⁹-bradykinin (kinin B₁ receptor agonist). This suggests that MAPK- and NF-κB-dependent kinin receptor upregulation can provide a novel option for treatment of AHR in asthmatic as well as in other inflammatory airway diseases.
Collapse
Affiliation(s)
- Yaping Zhang
- Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, Shaanxi 710021, People's Republic of China
| | | | | | | |
Collapse
|