1
|
Brak HH, Thielman NRJ. Norepinephrine mediates adrenergic receptor transcription and oncogenic gene expression in pancreatic ductal adenocarcinoma. Adv Biol Regul 2025; 97:101097. [PMID: 40349604 DOI: 10.1016/j.jbior.2025.101097] [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: 04/11/2025] [Accepted: 05/05/2025] [Indexed: 05/14/2025]
Abstract
Adrenergic stimulation of β2 receptors has shown to increase pancreatic ductal adenocarcinoma (PDAC) proliferation and migration in an in vitro setting; however, the role of α1 receptors in these adrenergic signaling pathways is unclear. Previous research has suggested that the MAPK signaling pathway is upregulated in response to β-adrenergic signaling, but other signaling pathways and downstream targets of mutant KRAS have yet to be investigated. This study investigates the role of adrenergic signaling through α1 and β-receptors in two human-derived PDAC cell lines, examining proliferation, wound healing, and protein expression after treatment with norepinephrine (NE) and in the presence of β and α1-receptor antagonism. Using RT-qPCR, the expression of adrenergic receptors and downstream KRAS effector proteins was evaluated. We found that NE has varying effects on proliferation and wound healing in different PDAC cell lines. Moreover, adrenergic receptor expression is under negative feedback control through α1 signaling in both cell lines. Furthermore, NE decreases expression of MMP9 while also affecting expression of VIM, CCND1, mTOR, and rhoA. We demonstrate genotype dependent effects of adrenergic stimulation on downstream molecular signaling pathways in PDAC that are important for oncogenicity. Based on our findings, genotypic characterization of cell signaling pathways in PDAC may aid further research in effective therapeutics for PDAC.
Collapse
Affiliation(s)
- Henry H Brak
- Lake Erie College of Osteopathic Medicine, Erie, PA, 16509, USA
| | | |
Collapse
|
2
|
Dong J, Che J, Wu Y, Deng Y, Jiang X, He Z, Zhang J. Dexmedetomidine promotes colorectal cancer progression via Piwil2 signaling. Cell Oncol (Dordr) 2024; 47:1459-1474. [PMID: 38592610 DOI: 10.1007/s13402-024-00944-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/27/2024] [Indexed: 04/10/2024] Open
Abstract
PURPOSE α2-adrenoceptor agonist dexmedetomidine (DEX) has been reported to promote tumorigenesis. Stem-cell protein Piwil2 is associated with cancer progression. Whether Piwil2 plays a role in tumor-promoting effects of DEX is unknown. METHODS We examined the expression of Piwil2 in human colorectal cancer (CRC) cell lines with/without DEX treatment. We also studied the roles of Piwil2 in proliferation, invasion, migration, as well as expressions of epithelial-mesenchymal transition (EMT)-related proteins in DEX-treated in vitro and in vivo CRC models. And the experiments with genetic and pharmacological treatments were conducted to investigate the underlying molecular mechanism. RESULTS RNA-sequencing (RNA-seq) analysis found Piwil2 is one of most upregulated genes upon DEX treatment in CRC cells. Furthermore, Piwil2 protein levels significantly increased in DEX-treated CRC cancer cells, which promoted proliferation, invasion, and migration in both CRC cell lines and human tumor xenografts model. Mechanistically, DEX increased nuclear factor E2-related factor 2 (Nrf2) expression, which enhanced Piwil2 transcription via binding to its promoter. Furthermore, in vitro experiments with Piwil2 knockdown or Siah2 inhibition indicated that DEX promoted EMT process and tumorigenesis through Siah2/PHD3/HIF1α pathway. The experiments with another α2-adrenoceptor agonist Brimonidine and antagonists yohimbine and atipamezole also suggested the role of Piwil2 signaling in tumor-promoting effects via an α2 adrenoceptor-dependent manner. CONCLUSION DEX promotes CRC progression may via activating α2 adrenoceptor-dependent Nrf2/Piwil2/Siah2 pathway and thus EMT process. Our work provides a novel insight into the mechanism underlying tumor-promoting effects of α2-adrenoceptor agonists.
Collapse
Affiliation(s)
- Jing Dong
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, No. 270 Dong'An Road, Xuhui District, Shanghai, 200032, P. R. China
| | - Ji Che
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, No. 270 Dong'An Road, Xuhui District, Shanghai, 200032, P. R. China
| | - Yuanyuan Wu
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, No. 270 Dong'An Road, Xuhui District, Shanghai, 200032, P. R. China
| | - Yixu Deng
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, No. 270 Dong'An Road, Xuhui District, Shanghai, 200032, P. R. China
| | - Xuliang Jiang
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, No. 270 Dong'An Road, Xuhui District, Shanghai, 200032, P. R. China
| | - Zhiyong He
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, No. 270 Dong'An Road, Xuhui District, Shanghai, 200032, P. R. China
| | - Jun Zhang
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, No. 270 Dong'An Road, Xuhui District, Shanghai, 200032, P. R. China.
- Department of Oncology, Shanghai Medical College, Fudan University, No. 270 Dong'An Road, Xuhui District, Shanghai, 200032, P. R. China.
| |
Collapse
|
3
|
Lim L, Kim H, Jeong J, Han SH, Yu YB, Song H. Yohimbine Inhibits PDGF-Induced Vascular Smooth Muscle Cell Proliferation and Migration via FOXO3a Factor. Int J Mol Sci 2024; 25:6899. [PMID: 39000009 PMCID: PMC11240894 DOI: 10.3390/ijms25136899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 06/16/2024] [Accepted: 06/21/2024] [Indexed: 07/14/2024] Open
Abstract
Yohimbine (YHB) has been reported to possess anti-inflammatory, anticancer, and cardiac function-enhancing properties. Additionally, it has been reported to inhibit the proliferation, migration, and neointimal formation of vascular smooth muscle cells (VSMCs) induced by platelet-derived growth factor (PDGF) stimulation by suppressing the phospholipase C-gamma 1 pathway. However, the transcriptional regulatory mechanism of YHB controlling the behavior of VSMCs is not fully understood. In this study, YHB downregulated the expression of cell cycle regulatory proteins, such as proliferating cell nuclear antigen (PCNA), cyclin D1, cyclin-dependent kinase 4 (CDK4), and cyclin E, by modulating the transcription factor FOXO3a in VSMCs induced by PDGF. Furthermore, YHB decreased p-38 and mTOR phosphorylation in a dose-dependent manner. Notably, YHB significantly reduced the phosphorylation at Y397 and Y925 sites of focal adhesion kinase (FAK), and this effect was greater at the Y925 site than Y397. In addition, the expression of paxillin, a FAK-associated protein known to bind to the Y925 site of FAK, was significantly reduced by YHB treatment in a dose-dependent manner. A pronounced reduction in the migration and proliferation of VSMCs was observed following co-treatment of YHB with mTOR or p38 inhibitors. In conclusion, this study shows that YHB inhibits the PDGF-induced proliferation and migration of VSMCs by regulating the transcription factor FOXO3a and the mTOR/p38/FAK signaling pathway. Therefore, YHB may be a potential therapeutic candidate for preventing and treating cardiovascular diseases such as atherosclerosis and vascular restenosis.
Collapse
MESH Headings
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/drug effects
- Forkhead Box Protein O3/metabolism
- Cell Proliferation/drug effects
- Cell Movement/drug effects
- Animals
- Platelet-Derived Growth Factor/metabolism
- Platelet-Derived Growth Factor/pharmacology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/drug effects
- Phosphorylation/drug effects
- Yohimbine/pharmacology
- Rats
- Signal Transduction/drug effects
- TOR Serine-Threonine Kinases/metabolism
- p38 Mitogen-Activated Protein Kinases/metabolism
- Focal Adhesion Kinase 1/metabolism
- Cells, Cultured
- Paxillin/metabolism
- Rats, Sprague-Dawley
- Male
Collapse
Affiliation(s)
- Leejin Lim
- Advanced Cancer Controlling Research Center, Chosun University, Gwangju 61452, Republic of Korea
| | - Hyeonhwa Kim
- Department of Biomedical Sciences, Chosun University Graduate School, Gwangju 61452, Republic of Korea
| | - Jihye Jeong
- Department of Biomedical Sciences, Chosun University Graduate School, Gwangju 61452, Republic of Korea
| | - Sung Hee Han
- Institute of Human Behavior & Genetics, Biomedical Research Center, Korea University, Seoul 02841, Republic of Korea
| | - Young-Bob Yu
- Department of Paramedicine, Nambu University, Gwangju 62271, Republic of Korea
| | - Heesang Song
- Advanced Cancer Controlling Research Center, Chosun University, Gwangju 61452, Republic of Korea
- Department of Biomedical Sciences, Chosun University Graduate School, Gwangju 61452, Republic of Korea
- Department of Biochemistry and Molecular Biology, Chosun University School of Medicine, Gwangju 61452, Republic of Korea
| |
Collapse
|
4
|
Fisher JL, Wilk EJ, Oza VH, Gary SE, Howton TC, Flanary VL, Clark AD, Hjelmeland AB, Lasseigne BN. Signature reversion of three disease-associated gene signatures prioritizes cancer drug repurposing candidates. FEBS Open Bio 2024; 14:803-830. [PMID: 38531616 PMCID: PMC11073506 DOI: 10.1002/2211-5463.13796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Revised: 03/13/2024] [Accepted: 03/14/2024] [Indexed: 03/28/2024] Open
Abstract
Drug repurposing is promising because approving a drug for a new indication requires fewer resources than approving a new drug. Signature reversion detects drug perturbations most inversely related to the disease-associated gene signature to identify drugs that may reverse that signature. We assessed the performance and biological relevance of three approaches for constructing disease-associated gene signatures (i.e., limma, DESeq2, and MultiPLIER) and prioritized the resulting drug repurposing candidates for four low-survival human cancers. Our results were enriched for candidates that had been used in clinical trials or performed well in the PRISM drug screen. Additionally, we found that pamidronate and nimodipine, drugs predicted to be efficacious against the brain tumor glioblastoma (GBM), inhibited the growth of a GBM cell line and cells isolated from a patient-derived xenograft (PDX). Our results demonstrate that by applying multiple disease-associated gene signature methods, we prioritized several drug repurposing candidates for low-survival cancers.
Collapse
Affiliation(s)
- Jennifer L. Fisher
- Department of Cell, Developmental and Integrative Biology, Heersink School of MedicineThe University of Alabama at BirminghamALUSA
| | - Elizabeth J. Wilk
- Department of Cell, Developmental and Integrative Biology, Heersink School of MedicineThe University of Alabama at BirminghamALUSA
| | - Vishal H. Oza
- Department of Cell, Developmental and Integrative Biology, Heersink School of MedicineThe University of Alabama at BirminghamALUSA
| | - Sam E. Gary
- Department of Cell, Developmental and Integrative Biology, Heersink School of MedicineThe University of Alabama at BirminghamALUSA
| | - Timothy C. Howton
- Department of Cell, Developmental and Integrative Biology, Heersink School of MedicineThe University of Alabama at BirminghamALUSA
| | - Victoria L. Flanary
- Department of Cell, Developmental and Integrative Biology, Heersink School of MedicineThe University of Alabama at BirminghamALUSA
| | - Amanda D. Clark
- Department of Cell, Developmental and Integrative Biology, Heersink School of MedicineThe University of Alabama at BirminghamALUSA
| | - Anita B. Hjelmeland
- Department of Cell, Developmental and Integrative Biology, Heersink School of MedicineThe University of Alabama at BirminghamALUSA
| | - Brittany N. Lasseigne
- Department of Cell, Developmental and Integrative Biology, Heersink School of MedicineThe University of Alabama at BirminghamALUSA
| |
Collapse
|
5
|
Priyanka HP, Pratap UP, Nair RS, Vasantharekha R, ThyagaRajan S. Estrogen-receptor status determines differential regulation of α1- and α2-adrenoceptor-mediated cell survival, angiogenesis, and intracellular signaling responses in breast cancer cell lines. Med Oncol 2024; 41:92. [PMID: 38526769 DOI: 10.1007/s12032-024-02322-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 02/01/2024] [Indexed: 03/27/2024]
Abstract
Psychosocial stress promotes cancer pathogenesis involving angiogenesis through alterations in neuroendocrine-immune functions that may involve adrenoceptor (AR)-dependent signaling mechanisms in the brain, lymphoid organs, and cancerous cells. Various concentrations of α1- and α2- AR-specific agonists and antagonists were incubated in vitro with estrogen receptor-positive (ER +) MCF-7, and ER (-) MDA MB-231 cells to examine the secretions of VEGF-A, VEGF-C, and nitric oxide (NO), and expression of signaling molecules- p-ERK, p-CREB, and p-Akt on the proliferation of breast cancer cell lines. Cellular proliferation, VEGF-A and NO secretion, expression of p-ERK, p-CREB, and p-Akt were enhanced in MCF-7 cells treated with α1-AR agonist while VEGF-C secretion alone was enhanced in MDA MB-231 cells. Treatment of MCF-7 and MDA MB-231 cells with α2- AR agonist similarly enhanced proliferation and decreased NO production and p-CREB expression while VEGF-C secretion was decreased in MCF-7 cells and p-Akt expression was decreased in MDA MB-231 cells. α1-AR inhibition reversed cellular proliferation and VEGF-A secretion by MCF-7 cells while α2-AR inhibition reversed the proliferation of MCF-7 and MDA MB-231 cells and VEGF-C secretion by MCF-7 cells. Taken together, breast cancer pathogenesis may be influenced by distinct α-AR-mediated signaling mechanisms on angiogenesis and lymphangiogenesis that are dependent on estrogen receptor status.
Collapse
Grants
- BT/PR9199/Med/30/12/2007 Department of Bio-Technology, Government of India, New Delhi.
- BT/PR9199/Med/30/12/2007 Department of Bio-Technology, Government of India, New Delhi.
- BT/PR9199/Med/30/12/2007 Department of Bio-Technology, Government of India, New Delhi.
- BT/PR9199/Med/30/12/2007 Department of Bio-Technology, Government of India, New Delhi.
- BT/PR9199/Med/30/12/2007 Department of Bio-Technology, Government of India, New Delhi.
Collapse
Affiliation(s)
- Hannah P Priyanka
- Institute of Advanced Research in Health Sciences, Tamil Nadu Government Multi Super Speciality Hospital, Omandurar Government Estate, Chennai, Tamil Nadu, India
- Integrative Medicine Laboratory, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Uday P Pratap
- Integrative Medicine Laboratory, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Rahul S Nair
- Institute of Advanced Research in Health Sciences, Tamil Nadu Government Multi Super Speciality Hospital, Omandurar Government Estate, Chennai, Tamil Nadu, India
| | - Ramasamy Vasantharekha
- Integrative Medicine Laboratory, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India.
| | - Srinivasan ThyagaRajan
- Institute of Advanced Research in Health Sciences, Tamil Nadu Government Multi Super Speciality Hospital, Omandurar Government Estate, Chennai, Tamil Nadu, India
| |
Collapse
|
6
|
Jabir NR, Firoz CK, Zughaibi TA, Alsaadi MA, Abuzenadah AM, Al-Asmari AI, Alsaieedi A, Ahmed BA, Ramu AK, Tabrez S. A literature perspective on the pharmacological applications of yohimbine. Ann Med 2022; 54:2861-2875. [PMID: 36263866 PMCID: PMC9590431 DOI: 10.1080/07853890.2022.2131330] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Introduction: Phytochemicals have garnered much attention because they are useful in managing several human diseases. Yohimbine is one such phytochemical with significant pharmacological potential and could be exploited for research by medicinal chemists. It is an indole alkaloid obtained from various natural/synthetic sources.Aims and Results: The research on yohimbine started early, and its use as a stimulant and aphrodisiac by humans has been reported for a long time. The pharmacological activity of yohimbine is mediated by the combined action of the central and peripheral nervous systems. It selectively blocks the pre and postsynaptic α2-adrenergic receptors and has a moderate affinity for α1 and α2 subtypes. Yohimbine also binds to other behaviourally relevant monoaminergic receptors in the following order: α-2 NE > 5HT-1A>, 5HT-1B > 1-D > D3 > D2 receptors.Conclusion: The current review highlights some significant findings that contribute to developing yohimbine-based drugs. It also highlights the therapeutic potential of yohimbine against selected human diseases. However, further research is recommended on the pharmacokinetics, molecular mechanisms, and drug safety requirements using well-designed randomized clinical trials to produce yohimbine as a pharmaceutical agent for human use.Key MessagesYohimbine is a natural indole alkaloid with significant pharmacological potential.Humans have used it as a stimulant and aphrodisiac from a relatively early time.It blocks the pre- and postsynaptic α2-adrenergic receptors that could be exploited for managing erectile dysfunction, myocardial dysfunction, inflammatory disorders, and cancer.
Collapse
Affiliation(s)
- Nasimudeen R Jabir
- Department of Biochemistry and Biotechnology, Centre for Research and Development, PRIST University, Vallam, Thanjavur, India
| | - Chelapram K Firoz
- Department of Medical Laboratory Technology, MIMS College of Allied Health Sciences, ASTER MIMS Academy, Malappuram, Kerala University of Health Sciences, Kerala, India
| | - Torki A Zughaibi
- Toxicology and Forensic Science Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohammed Abdullah Alsaadi
- Hematology Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Adel M Abuzenadah
- Toxicology and Forensic Science Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ahmed Ibrahim Al-Asmari
- Toxicology and Forensic Science Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia.,Laboratory Department, King Abdul-Aziz Hospital, Ministry of Health, Jeddah, Saudi Arabia
| | - Ahdab Alsaieedi
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia.,Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Bakrudeen Ali Ahmed
- Department of Biochemistry and Biotechnology, Centre for Research and Development, PRIST University, Vallam, Thanjavur, India
| | - Arun Kumar Ramu
- Department of Biochemistry and Biotechnology, Centre for Research and Development, PRIST University, Vallam, Thanjavur, India
| | - Shams Tabrez
- Toxicology and Forensic Science Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
7
|
Banik A, Ahmed SR, Sajib EH, Deb A, Sinha S, Azim KF. Identification of potential inhibitory analogs of metastasis tumor antigens (MTAs) using bioactive compounds: revealing therapeutic option to prevent malignancy. Mol Divers 2022; 26:2473-2502. [PMID: 34743299 DOI: 10.1007/s11030-021-10345-w] [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: 02/19/2021] [Accepted: 10/24/2021] [Indexed: 12/31/2022]
Abstract
The deeper understanding of metastasis phenomenon and detection of drug targets could be a potential approach to minimize cancer mortality. In this study, attempts were taken to unmask novel therapeutics to prevent metastasis and cancer progression. Initially, we explored the physiochemical, structural and functional insights of three metastasis tumor antigens (MTAs) and evaluated some plant-based bioactive compounds as potent MTA inhibitors. From 50 plant metabolites screened, isoflavone, gingerol, citronellal and asiatic acid showed maximum binding affinity with all three MTA proteins. The ADME analysis detected no undesirable toxicity that could reduce the drug likeness properties of top plant metabolites. Moreover, molecular dynamics studies revealed that the complexes were stable and showed minimum fluctuation at molecular level. We further performed ligand-based virtual screening to identify similar drug molecules using a large collection of 376,342 compounds from DrugBank. The results suggested that several structural analogs (e.g., tramadol, nabumetone, DGLA and hydrocortisone) may act as agonist to block the MTA proteins and inhibit cancer progression at early stage. The study could be useful to develop effective medications against cancer metastasis in future. Due to encouraging results, we highly recommend further in vitro and in vivo trials for the experimental validation of the findings.
Collapse
Affiliation(s)
- Anik Banik
- Faculty of Biotechnology and Genetic Engineering, Sylhet Agricultural University, Sylhet, 3100, Bangladesh
- Department of Plant and Environmental Biotechnology, Sylhet Agricultural University, Sylhet, 3100, Bangladesh
| | - Sheikh Rashel Ahmed
- Faculty of Biotechnology and Genetic Engineering, Sylhet Agricultural University, Sylhet, 3100, Bangladesh
- Department of Plant and Environmental Biotechnology, Sylhet Agricultural University, Sylhet, 3100, Bangladesh
| | - Emran Hossain Sajib
- Faculty of Biotechnology and Genetic Engineering, Sylhet Agricultural University, Sylhet, 3100, Bangladesh
| | - Anamika Deb
- Faculty of Biotechnology and Genetic Engineering, Sylhet Agricultural University, Sylhet, 3100, Bangladesh
| | - Shiuly Sinha
- Faculty of Biotechnology and Genetic Engineering, Sylhet Agricultural University, Sylhet, 3100, Bangladesh
| | - Kazi Faizul Azim
- Department of Microbial Biotechnology, Sylhet Agricultural University, Sylhet, 3100, Bangladesh.
- Faculté de Pharmacie, Université de Tours, 37200, Tours, France.
| |
Collapse
|
8
|
Jabir NR, Khan MS, Alafaleq NO, Naz H, Ahmed BA. Anticancer potential of yohimbine in drug-resistant oral cancer KB-ChR-8-5 cells. Mol Biol Rep 2022; 49:9565-9573. [PMID: 35970968 DOI: 10.1007/s11033-022-07847-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 08/05/2022] [Indexed: 11/26/2022]
Abstract
BACKGROUND The demand for environmentally friendly and cost-effective plant-based products for the development of cancer therapeutics has been increasing. Yohimbine (α2-adrenergic receptor antagonist) is a stimulant and aphrodisiac used to improve erectile dysfunction. In this study, we aimed to evaluate the anticancer potential of yohimbine in drug-resistant oral cancer KB-ChR-8-5 cells using different biomolecular techniques. METHODS We estimated the anticancer efficacy of yohimbine using different assays, such as MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) cell cytotoxicity, cell morphology, cell apoptosis, reactive oxygen species (ROS) formation, and modulation in the mitochondrial membrane potential (MMP). RESULTS Yohimbine showed a dose-dependent increase in cytotoxicity with a 50% inhibitory concentration (IC50) of 44 µM against KB-ChR-8-5 cancer cell lines. Yohimbine treatment at 40 µM and 50 µM resulted in a considerable change in cell morphology, including shrinkage, detachment, membrane blebbing, and deformed shape. Moreover, at the dose of IC50 and above, a significant induction was observed in the generation of ROS and depolarization of MMP. The possible mechanisms of action of yohimbine underlying the dose-dependent increase in cytotoxicity may be due to the induction of apoptosis, ROS generation, and modulation of MMP. CONCLUSION Overall, yohimbine showed a significant anticancer potential against drug-resistant oral cancer KB-ChR-8-5 cells. Our study suggests that besides being an aphrodisiac, yohimbine can be used as a drug repurposing agent. However, more research is required in different in vitro and in vivo models to confirm the feasibility of yohimbine in clinics.
Collapse
Affiliation(s)
- Nasimudeen R Jabir
- Department of Biochemistry, Centre for Research and Development, PRIST University, Vallam, Thanjavur, Tamil Nadu, 613403, India
| | - Mohd Shahnawaz Khan
- Department of Biochemistry, College of Sciences, King Saud University, Riyadh, 11451, Saudi Arabia.
| | - Nouf Omar Alafaleq
- Department of Biochemistry, College of Sciences, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Huma Naz
- Department of Medicine, University of Missouri, Columbia, MO, 65201, USA
| | - Bakrudeen Ali Ahmed
- Department of Biochemistry, Centre for Research and Development, PRIST University, Vallam, Thanjavur, Tamil Nadu, 613403, India.
| |
Collapse
|
9
|
Chiu CW, Hsieh CY, Yang CH, Tsai JH, Huang SY, Sheu JR. Yohimbine, an α2-Adrenoceptor Antagonist, Suppresses PDGF-BB-Stimulated Vascular Smooth Muscle Cell Proliferation by Downregulating the PLCγ1 Signaling Pathway. Int J Mol Sci 2022; 23:ijms23148049. [PMID: 35887391 PMCID: PMC9324260 DOI: 10.3390/ijms23148049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 07/13/2022] [Accepted: 07/14/2022] [Indexed: 11/22/2022] Open
Abstract
Yohimbine (YOH) has antiproliferative effects against breast cancer and pancreatic cancer; however, its effects on vascular proliferative diseases such as atherosclerosis remain unknown. Accordingly, we investigated the inhibitory mechanisms of YOH in vascular smooth muscle cells (VSMCs) stimulated by platelet-derived growth factor (PDGF)-BB, a major mitogenic factor in vascular diseases. YOH (5–20 μM) suppressed PDGF-BB-stimulated a mouse VSMC line (MOVAS-1 cell) proliferation without inducing cytotoxicity. YOH also exhibited antimigratory effects and downregulated matrix metalloproteinase-2 and -9 expression in PDGF-BB-stimulated MOVAS-1 cells. It also promoted cell cycle arrest in the initial gap/first gap phase by upregulating p27Kip1 and p53 expression and reducing cyclin-dependent kinase 2 and proliferating cell nuclear antigen expression. We noted phospholipase C-γ1 (PLCγ1) but not ERK1/2, AKT, or p38 kinase phosphorylation attenuation in YOH-modulated PDGF-BB-propagated signaling pathways in the MOVAS-1 cells. Furthermore, YOH still inhibited PDGF-BB-induced cell proliferation and PLCγ1 phosphorylation in MOVAS-1 cells with α2B-adrenergic receptor knockdown. YOH (5 and 10 mg/kg) substantially suppressed neointimal hyperplasia in mice subjected to CCA ligation for 21 days. Overall, our results reveal that YOH attenuates PDGF-BB-stimulated VSMC proliferation and migration by downregulating a α2B-adrenergic receptor–independent PLCγ1 pathway and reduces neointimal formation in vivo. Therefore, YOH has potential for repurposing for treating atherosclerosis and other vascular proliferative diseases.
Collapse
Affiliation(s)
- Chih-Wei Chiu
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
| | - Cheng-Ying Hsieh
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; (C.-Y.H.); (C.-H.Y.); (J.-H.T.)
| | - Chih-Hao Yang
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; (C.-Y.H.); (C.-H.Y.); (J.-H.T.)
| | - Jie-Heng Tsai
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; (C.-Y.H.); (C.-H.Y.); (J.-H.T.)
| | - Shih-Yi Huang
- School of Nutrition and Health Sciences, Taipei Medical University, Taipei 110, Taiwan
- Graduate Institute of Metabolism and Obesity Sciences, Taipei Medical University, Taipei 110, Taiwan
- Center for Reproductive Medicine & Sciences, Taipei Medical University Hospital, Taipei 110, Taiwan
- Correspondence: (S.-Y.H.); (J.-R.S.); Tel.: +886-2-2736-1661 (ext. 6543) (S.-Y.H.); +886-2-2736-1661 (ext. 3199) (J.-R.S.)
| | - Joen-Rong Sheu
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; (C.-Y.H.); (C.-H.Y.); (J.-H.T.)
- Correspondence: (S.-Y.H.); (J.-R.S.); Tel.: +886-2-2736-1661 (ext. 6543) (S.-Y.H.); +886-2-2736-1661 (ext. 3199) (J.-R.S.)
| |
Collapse
|
10
|
Fu T, Qin X, Ma Y, Yuan XY, Wu S, Ye X, Dang Y. Yohimbine hydrochloride inhibits skin melanin synthesis by regulating wnt/β-catenin and p38/MAPK signal pathways. J Dermatol Sci 2022; 107:17-23. [DOI: 10.1016/j.jdermsci.2022.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 05/31/2022] [Accepted: 06/30/2022] [Indexed: 10/17/2022]
|
11
|
Abuzenadah AM, Al-Sayes F, Mahafujul Alam SS, Hoque M, Karim S, Hussain IMR, Tabrez S. Identification of Potential Poly (ADP-Ribose) Polymerase-1 Inhibitors Derived from Rauwolfia serpentina: Possible Implication in Cancer Therapy. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:3787162. [PMID: 35368755 PMCID: PMC8967534 DOI: 10.1155/2022/3787162] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 02/21/2022] [Indexed: 12/18/2022]
Abstract
Poly (ADP-ribose) polymerase-1 (PARP-1) has been recognized as a prospective target for the development of novel cancer therapeutics. Several PARP-1 inhibitors are currently being considered for anticancer drug development and clinical investigation. Lately, natural compounds seem to be excellent alternative drug candidates for cancer treatment. Rauwolfia serpentina is a medicinal plant traditionally used in Indian subcontinents to treat various diseases. This study has been designed to identify the bioactive compounds derived from R. serpentina for possible binding and inhibition of PARP-1 using the molecular docking approach. Thirteen compounds were found to interact with the target with a binding affinity greater than the value of -9.0 kcal/mol. After screening the physicochemical properties, only 5 ligands (ajmalicine, yohimbine, isorauhimbine, rauwolscine, and 1,2-dihydrovomilenine) were found to obey all the parameters of Lipinski's rule of five, showed maximum drug-likeness, and possess no significant toxicity. These ligands displayed strong interactions with target PARP-1 via several hydrogen bonds and hydrophobic interactions. Therefore, these identified compounds derived from R. serpentina can be considered for drug development against cancer-targeting PARP-1.
Collapse
Affiliation(s)
- Adel M. Abuzenadah
- Department of Medical Laboratory Science, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Center of Excellence in Genomic Medicine Research (CEGMR), King Abdulaziz University, Jeddah, Saudi Arabia
| | - Fatin Al-Sayes
- Center of Excellence in Genomic Medicine Research (CEGMR), King Abdulaziz University, Jeddah, Saudi Arabia
| | | | - Mehboob Hoque
- Applied Bio-Chemistry Lab, Department of Biological Sciences, Aliah University, Kolkata, India
| | - Sajjad Karim
- Center of Excellence in Genomic Medicine Research (CEGMR), King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ibtessam M. R. Hussain
- Center of Excellence in Genomic Medicine Research (CEGMR), King Abdulaziz University, Jeddah, Saudi Arabia
| | - Shams Tabrez
- Department of Medical Laboratory Science, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
12
|
Mucke HA. Drug Repurposing Patent Applications October: December 2021. Assay Drug Dev Technol 2022. [DOI: 10.1089/adt.2022.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
13
|
Abuzenadah AM, Al-Sayes F, Mahafujul Alam SS, Hoque M, Karim S, Hussain IMR, Tabrez S. Elucidating Antiangiogenic Potential of Rauwolfia serpentina: VEGFR-2 Targeting-Based Molecular Docking Study. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:6224666. [PMID: 35198035 PMCID: PMC8860507 DOI: 10.1155/2022/6224666] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 01/17/2022] [Indexed: 12/13/2022]
Abstract
Angiogenesis plays a critical role in tumorigenesis as it provides the necessary blood supply to the newly grown solid tumor. It helps maintain the tumor microenvironment, promotes tumor development, progression, and metastasis. The vascular epithelial growth factor (VEGF), interacting with the tyrosine kinase receptor VEGFR-2 on endothelial cells, exerts its proangiogenic activity. Hence, targeting the VEGFR-2 signaling is considered a promising strategy to inhibit angiogenesis and thus cancer treatment. This study aims to identify the bioactive compounds derived from the medicinal herb Rauwolfia serpentina that effectively binds with VEGFR-2. The bioactive compounds of R. serpentina were first screened for their physicochemical properties using the DataWarrior program (version 5.5.0). Finally, 17 compounds that obeyed Lipinski's rule of five and showed good drug-likeness were selected for molecular docking studies. Molecular docking results showed that the ligands ajmalicidine, 1, 2-dihydrovomilenine, rauwolscine, yohimbine, ajmaline, and papaverine interact strongly with the target VEGFR-2 receptor. Hydrogen bonds and hydrophobic interactions stabilized the interactions of these compounds with VEGFR-2. These compounds showed favourable drug-like properties and possess no significant toxicity. Therefore, the findings of this study indicate that the compounds derived from R. serpentina can be considered for the development of antiangiogenic drug candidates by targeting VEGFR-2.
Collapse
Affiliation(s)
- Adel M. Abuzenadah
- Department of Medical Laboratory Science, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Center of Excellence in Genomic Medicine Research (CEGMR), King Abdulaziz University, Jeddah, Saudi Arabia
| | - Fatin Al-Sayes
- Center of Excellence in Genomic Medicine Research (CEGMR), King Abdulaziz University, Jeddah, Saudi Arabia
| | | | - Mehboob Hoque
- Applied Bio-Chemistry Lab, Department of Biological Sciences, Aliah University, Kolkata, India
| | - Sajjad Karim
- Center of Excellence in Genomic Medicine Research (CEGMR), King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ibtessam M. R. Hussain
- Center of Excellence in Genomic Medicine Research (CEGMR), King Abdulaziz University, Jeddah, Saudi Arabia
| | - Shams Tabrez
- Department of Medical Laboratory Science, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
14
|
Zhao Y, Zhang Y, Li Y, Yang M, Yuan J, Cao Y, Xu L, Ma X, Lin S, An J, Wang S. Yohimbine hydrochloride inhibits benign prostatic hyperplasia by downregulating steroid 5α-reductase type 2. Eur J Pharmacol 2021; 908:174334. [PMID: 34265299 DOI: 10.1016/j.ejphar.2021.174334] [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: 01/26/2021] [Revised: 07/02/2021] [Accepted: 07/11/2021] [Indexed: 10/20/2022]
Abstract
Benign prostatic hyperplasia (BPH) is a frequently encountered disease in older men that affects sexual function and is capable of causing lower urinary tract dysfunction. Unfortunately, current treatment options for BPH primarily seek to address the lower urinary tract dysfunction aspect of the disease and do not improve sexual function. Yohimbine has been effectively used for decades to treat erectile dysfunction. Therefore, the objective of this study was to evaluate the inhibitory effect of yohimbine on BPH and explore the associated underlying mechanisms. Thirty-six rats were randomly divided into the control, BPH, finasteride (1 mg/kg), and yohimbine (2, 4, and 8 mg/kg) groups. Except for the rats in the control group, those in the other groups were subcutaneously injected testosterone propionate (5 mg/kg/day) daily for a period of 4 weeks to establish BPH models. They were also administration the corresponding drug daily for a period of 6 weeks. After the treatments, in addition to determining prostate wet weight and index, the histopathological status of the prostate was observed, and the levels of testosterone, dihydrotestosterone, prostatic acid phosphatase, the prostate-specific antigen, proliferating cell nuclear antigen, and steroid 5α-reductase were determined. Specifically, the administration of 2, 4, and 8 mg/kg yohimbine inhibited prostatic index increase by 46.7, 55.1, and 69.3%, respectively, in BHP rats. Further, yohimbine significantly reduced the levels of dihydrotestosterone, prostatic acid phosphatase, prostate-specific antigen, proliferating cell nuclear antigen, and steroid 5α-reductase, suggesting that it exerts beneficial effects against BPH by modulating the steroid 5α-reductase pathway.
Collapse
Affiliation(s)
- Yani Zhao
- Xi'an Hospital of Traditional Chinese Medicine, Xi'an, 710021, Shaanxi, China.
| | - Yan Zhang
- Xi'an Hospital of Traditional Chinese Medicine, Xi'an, 710021, Shaanxi, China
| | - Yao Li
- Key Laboratory Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Science and Medicine, Northwest University, Xi'an, 710069, Shaanxi, China
| | - Min Yang
- Xi'an Hospital of Traditional Chinese Medicine, Xi'an, 710021, Shaanxi, China
| | - Jiani Yuan
- Air Force Hospital of Western Theater Command, Chengdu, 610000, Sichuan, China
| | - Yu Cao
- Department of Chinese Materia Medica and Natural Medicines, School of Pharmacy, The Air Force Medical University, Xi'an, 710032, Shaanxi, China
| | - Lu Xu
- Department of Chinese Materia Medica and Natural Medicines, School of Pharmacy, The Air Force Medical University, Xi'an, 710032, Shaanxi, China
| | - Xuexinyu Ma
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, 712046, Shaanxi, China
| | - Sisong Lin
- Key Laboratory Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Science and Medicine, Northwest University, Xi'an, 710069, Shaanxi, China
| | - Junming An
- Xi'an Hospital of Traditional Chinese Medicine, Xi'an, 710021, Shaanxi, China.
| | - Siwang Wang
- Key Laboratory Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Science and Medicine, Northwest University, Xi'an, 710069, Shaanxi, China.
| |
Collapse
|
15
|
A mononuclear PdII complex with Naphcon; crystal structure, experimental and computational studies of the interaction with DNA/BSA and evaluation of anticancer activity. Polyhedron 2021. [DOI: 10.1016/j.poly.2021.115333] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
16
|
Yang H, Poznik M, Tang S, Xue P, Du L, Liu C, Chen X, Chruma JJ. Synthesis of Conformationally Liberated Yohimbine Analogues and Evaluation of Cytotoxic Activity. ACS OMEGA 2021; 6:19291-19303. [PMID: 34337266 PMCID: PMC8320076 DOI: 10.1021/acsomega.1c02784] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 07/07/2021] [Indexed: 05/09/2023]
Abstract
A modular synthetic approach to strategically unique structural analogues of the alkaloid yohimbine is reported. The overall synthetic strategy couples the transition-metal-catalyzed decarboxylative allylation of 2,2-diphenylglycinate imino esters with a scandium triflate-mediated highly endo-selective intramolecular Diels-Alder (IMDA) cycloaddition to generate a small collection of de-rigidified yohimbine analogues lacking the ethylene linkage between the indole and decahydroisoquinoline units. One compound generated in this study contains an unprecedented pentacyclic urea core and appears to demonstrate increased cytotoxicity against the gastric cancer cell line SGC-7901 in comparison to a pancreatic cancer cell line (PATU-8988) and a normal human gastric mucosal cell line (GES-1).
Collapse
Affiliation(s)
- Han Yang
- Key
Laboratory of Green Chemistry & Technology (MOE), College of Chemistry
and Sino-British Materials Research Institute, College of Physical
Sciences & Technology, Sichuan University, Chengdu, Sichuan 610064, P. R. China
| | - Michal Poznik
- Key
Laboratory of Green Chemistry & Technology (MOE), College of Chemistry
and Sino-British Materials Research Institute, College of Physical
Sciences & Technology, Sichuan University, Chengdu, Sichuan 610064, P. R. China
| | - Shaojian Tang
- School
of Pharmacy, Weifang Medical University, No. 7166, Baotong West Road, Weifang, Shandong 261053, P. R. China
| | - Peng Xue
- School
of Public Health, Weifang Medical University, No. 7166, Baotong West Road, Weifang, Shandong 261053, P. R. China
| | - Lidong Du
- School
of Clinical Medicine, Weifang Medical University, No. 7166, Baotong West Road, Weifang, Shandong 261053, P. R. China
| | - Chenlu Liu
- Key
Laboratory of Green Chemistry & Technology (MOE), College of Chemistry
and Sino-British Materials Research Institute, College of Physical
Sciences & Technology, Sichuan University, Chengdu, Sichuan 610064, P. R. China
| | - Xiaochuan Chen
- Key
Laboratory of Green Chemistry & Technology (MOE), College of Chemistry
and Sino-British Materials Research Institute, College of Physical
Sciences & Technology, Sichuan University, Chengdu, Sichuan 610064, P. R. China
| | - Jason J. Chruma
- Key
Laboratory of Green Chemistry & Technology (MOE), College of Chemistry
and Sino-British Materials Research Institute, College of Physical
Sciences & Technology, Sichuan University, Chengdu, Sichuan 610064, P. R. China
- Department
of Chemistry, University of Virginia, McCormick Road, PO Box 400319, Charlottesville, Virginia 22903-4319, United States
| |
Collapse
|
17
|
Anitha R, Subashini R, Kannayiram G, Gayathri D. Chronic Inflammatory-Modulating Potential of Cassia auriculata with Proinflammatory Cytokine IL-1beta and Its Anticancer Effect on Lung Cancer Cell Line. Anticancer Agents Med Chem 2021; 21:343-354. [PMID: 32781968 DOI: 10.2174/1871520620666200811111114] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 06/25/2020] [Accepted: 07/09/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Inflammation is a key element in tumor progression, over time, persistent inflammation causes damage to DNA and leads to cancer. The relationship between chronic inflammation and tumor development is well established, blocking of which can help in cancer prevention and treatment in the future. OBJECTIVE Hence, with this background, the present study aims to evaluate the anti-inflammatory and anticancer potential of Cassia auriculata (CA) solvent fractions through in silico and in vitro means, respectively. METHODS Generally, inflammatory mediators play a key task in chronic inflammation, following its inflection was chosen for their interactions with nine structurally varied phytoconstituents of CA identified through GCMS. The ethanolic extract of CA was assessed for its apoptotic effects on A549 lung cancer cells by 3-(4,5- dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay, JC-10 staining, DNA fragmentation assay and quantitative Real-Time Polymerase Chain Reaction (qRT-PCR). RESULTS The interactions between bioactive components and target protein revealed that important molecules like 5,7-dihydroxy-2-[2-nethoxyphenyl]- 4H-1-Benzopyran-4-one, a flavonoid, and three other components can bind target interleukin 1-beta associated with lung cancer. In vitro data also confirmed that the diverse active components of CA extract might follow the intrinsic mitochondrial pathway to provoke cancer cell death. CONCLUSION Hence, these findings strongly propose that Cassia auriculata (CA) solvent fractions could be exploited in the future to design ligands for obtaining novel leads for treating chronic inflammation linked with lung cancer, and also the extracts of CA can be recommended as a potential agent for lung cancer chemotherapy.
Collapse
Affiliation(s)
- Rajagopal Anitha
- Department of Biomedical Engineering, SSN College of Engineering, Kalavakkam 603110, Tamil Nadu, India
| | - Rajakannu Subashini
- Department of Biomedical Engineering, SSN College of Engineering, Kalavakkam 603110, Tamil Nadu, India
| | - Gomathi Kannayiram
- Department of Biotechnology, Dr. MGR Educational and Research Institute, Maduravoyal, Chennai, Tamil Nadu 600095, India
| | - Dasararaju Gayathri
- Department of Crystallography and Biophysics, University of Madras, Chepauk, Chennai, Tamil Nadu 600005, India
| |
Collapse
|
18
|
Wang W, Guo X, Dan H. α2A-Adrenergic Receptor Inhibits the Progression of Cervical Cancer Through Blocking PI3K/AKT/mTOR Pathway. Onco Targets Ther 2020; 13:10535-10546. [PMID: 33116632 PMCID: PMC7574911 DOI: 10.2147/ott.s264409] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 08/28/2020] [Indexed: 12/19/2022] Open
Abstract
Objective The study aimed to investigate the effect of α2A-adrenergic receptor (ADRA2A) on cervical cancer and the potential mechanisms of ADRA2A on phosphatidylinositol 3′-kinase/protein kinase B/mammalian target of rapamycin (PI3K/Akt/mTOR) pathway in cervical cancer cells. Methods In our study, ADRA2A expression was evaluated by analyzing cervical cancer RNA sequencing dataset from the GEPIA. The prognostic values of ADRA2A were evaluated by Kaplan–Meier method using the Cancer Genome Atlas (TCGA) database data. In addition, the expression of ADRA2A in cervical cancer cell lines was detected by qRT-PCR and Western blot. Subsequently, the roles of ADRA2A on cell proliferation, apoptosis, migration, invasion and senescence in HeLa and SiHa cells were evaluated. Moreover, tumorigenesis in nude mice was used to investigate the role of ADRA2A in vivo. We also detected the expression changes of key factors in PI3K/Akt/mTOR pathway after overexpression and silencing of ADRA2A in HeLa and SiHa cells. Results ADRA2A expression was significantly downregulated in cervical cancer tissues and cell lines. The high expression of ADRA2A was significantly associated with a better prognosis in cervical cancer patients. ADRA2A overexpression significantly suppressed cell proliferation, migration and invasion, and promoted cell senescence and apoptosis in cervical cancer cells. On the contrary, silencing ADRA2A dramatically facilitated cell proliferation, migration and invasion, and inhibited cell senescence and apoptosis in cervical cancer cells. The expressions of p-PI3K, p-AKT and p-mTOR in cervical cancer cells were notably decreased by ADRA2A overexpression and increased by silencing ADRA2A. In addition, we also confirmed that ADRA2A overexpression could suppress the xenograft tumor growth in vivo. Conclusion Our study demonstrated that ADRA2A could suppress cell proliferation, migration and invasion, as well as promote cell senescence and apoptosis through inhibiting PI3K/Akt/mTOR pathway in cervical cancer.
Collapse
Affiliation(s)
- Weina Wang
- Health Management Center, Qingdao Sixth People's Hospital, Qingdao, Shandong 266011, People's Republic of China
| | - Xin Guo
- Department of Gynecology and Obstetrics, The People's Liberation Army Navy, The 971th Hospital, Qingdao, Shandong 266071, People's Republic of China
| | - Huiwen Dan
- Department of Gynecology and Obstetrics, The People's Liberation Army Navy, The 971th Hospital, Qingdao, Shandong 266071, People's Republic of China
| |
Collapse
|
19
|
Missair A, Cata JP, Votta-Velis G, Johnson M, Borgeat A, Tiouririne M, Gottumukkala V, Buggy D, Vallejo R, Marrero EBD, Sessler D, Huntoon MA, Andres JD, Casasola ODL. Impact of perioperative pain management on cancer recurrence: an ASRA/ESRA special article. Reg Anesth Pain Med 2019; 44:13-28. [PMID: 30640648 DOI: 10.1136/rapm-2018-000001] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Revised: 05/31/2018] [Accepted: 06/05/2018] [Indexed: 12/31/2022]
Abstract
Cancer causes considerable suffering and 80% of advanced cancer patients experience moderate to severe pain. Surgical tumor excision remains a cornerstone of primary cancer treatment, but is also recognized as one of the greatest risk factors for metastatic spread. The perioperative period, characterized by the surgical stress response, pharmacologic-induced angiogenesis, and immunomodulation results in a physiologic environment that supports tumor spread and distant reimplantation.In the perioperative period, anesthesiologists may have a brief and uniquewindow of opportunity to modulate the unwanted consequences of the stressresponse on the immune system and minimize residual disease. This reviewdiscusses the current research on analgesic therapies and their impact ondisease progression, followed by an evidence-based evaluation of perioperativepain interventions and medications.
Collapse
Affiliation(s)
- Andres Missair
- Department of Anesthesiology, Veterans Affairs Hospital, Miami, Florida, USA .,Department of Anesthesiology, University of Miami, Miami, Florida, USA
| | - Juan Pablo Cata
- Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Gina Votta-Velis
- Department of Anesthesiology, University of Illinois Hospital and Health Sciences System, Chicago, Illinois, USA
| | - Mark Johnson
- Department of Anesthesiology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Alain Borgeat
- Department of Anesthesiology, University of Zurich, Balgrist, Switzerland
| | - Mohammed Tiouririne
- Department of Anesthesiology, University of Virginia Health System, Charlottesville, Virginia, USA
| | - Vijay Gottumukkala
- Anesthesiology and Perioperative Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Donal Buggy
- Department of Anesthesiology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Ricardo Vallejo
- Department of Anesthesiology, Illinois Wesleyan University, Bloomington, Illinois, USA
| | - Esther Benedetti de Marrero
- Department of Anesthesiology, Veterans Affairs Hospital, Miami, Florida, USA.,Department of Anesthesiology, University of Miami, Miami, Florida, USA
| | - Dan Sessler
- Department of Anesthesiology and Pain Management, Cleveland Clinic, Cleveland, Ohio, USA
| | - Marc A Huntoon
- Department of Anesthesiology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Jose De Andres
- Department of Anesthesiology, General University Hospital, Valencia, Spain
| | - Oscar De Leon Casasola
- Department of Anesthesiology, University of Buffalo / Roswell Park Cancer Institute, Buffalo, New York, USA
| |
Collapse
|
20
|
Liu Y, Yu X, Zhuang J. Epinephrine Stimulates Cell Proliferation and Induces Chemoresistance in Myeloma Cells through the β-Adrenoreceptor in vitro. Acta Haematol 2017; 138:103-110. [PMID: 28848082 DOI: 10.1159/000478517] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 06/06/2017] [Indexed: 01/07/2023]
Abstract
OBJECTIVES To explore the effect of the β-adrenoreceptor signaling pathway on myeloma cells. METHODS The myeloma U266 cell line was treated with epinephrine and propranolol. Cell proliferation was analyzed by MTS assay. Apoptosis was detected by flow cytometry. The β-receptor subtype and the key enzyme of epinephrine were identified by reverse transcription polymerase chain reaction (RT-PCR). RESULTS Epinephrine (5-50 μM) promoted U266 cell growth in a dose-dependent manner and neutralized the inhibition effect of bortezomib (25 and 50 ng/mL) in vitro. Cell proliferation was inhibited by a β-receptor antagonist, propranolol, at a concentration of 50-200 μM. The proportions of early and late apoptotic cells were enhanced after treatment with propranolol. The expression of caspase 3/7, 8, and 9 was elevated in propranolol-treated myeloma cells. Both β1- and β2-adrenoceptor mRNAs were expressed in the U266 cell line. Key enzymes dopamine hydroxylase and tyrosinehydroxylase were identified in myeloma cells. CONCLUSIONS Our results reveal that epinephrine stimulates myeloma cell growth in vitro while the β-blocker propranolol has an antiproliferative effect, indicating that stress hormones may trigger the progression of myeloma.
Collapse
Affiliation(s)
- Yang Liu
- Department of Hematology, Peking Union Medical College Hospital, Beijing, China
| | | | | |
Collapse
|
21
|
Coelho M, Soares-Silva C, Brandão D, Marino F, Cosentino M, Ribeiro L. β-Adrenergic modulation of cancer cell proliferation: available evidence and clinical perspectives. J Cancer Res Clin Oncol 2017; 143:275-291. [PMID: 27709364 DOI: 10.1007/s00432-016-2278-1] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 09/21/2016] [Indexed: 12/30/2022]
Abstract
PURPOSE In this review, we aimed to present and discuss the available preclinical and epidemiological evidences regarding the modulation of cancer cell proliferation by β-adrenoceptors (β-AR), with a specific focus on the putative effects of β-blockers according to their pharmacological properties. METHODS A comprehensive review of the published literature was conducted, and the evidences concerning the involvement of β-AR in cancer as well as the possible role of β-blockers were selected and discussed. RESULTS The majority of reviewed studies show that: (1) All the cancer types express both β1- and β2-AR, with the exception of neuroblastoma only seeming to express β2-AR; (2) adrenergic agonists are able to increase proliferation of several types of cancers; (3) the proliferative effect seems to be mediated by both β1- and β2-AR; (4) binding to β-AR results in a cAMP transient flux which activates two major downstream effector systems: protein kinase A and EPAC and (5) β-blockers might be putative adjuvants for cancer treatment. CONCLUSIONS Overall, the reviewed studies show strong evidences that β-AR activation, through several intracellular mechanisms, modulate tumor cell proliferation suggesting β-blockers can be a feasible therapeutic approach to antagonize β-adrenergic response or have a protective effect per se. This review highlight the need for intensifying the research not only on the molecular mechanisms underlying the β-adrenergic influence in cancer, but also on the implications of biased agonism of β-blockers as potential antitumor agents.
Collapse
Affiliation(s)
- Marisa Coelho
- Department of Biochemistry, Faculty of Medicine, University of Porto, Porto, Portugal
- I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Center for Research in Medical Pharmacology, University of Insubria, Varese, Italy
| | - Cátia Soares-Silva
- Department of Biochemistry, Faculty of Medicine, University of Porto, Porto, Portugal
- I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Daniela Brandão
- Department of Biochemistry, Faculty of Medicine, University of Porto, Porto, Portugal
- Department of Medical Education and Simulation, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Franca Marino
- Center for Research in Medical Pharmacology, University of Insubria, Varese, Italy
| | - Marco Cosentino
- Center for Research in Medical Pharmacology, University of Insubria, Varese, Italy
| | - Laura Ribeiro
- Department of Biochemistry, Faculty of Medicine, University of Porto, Porto, Portugal.
- I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.
- Department of Medical Education and Simulation, Faculty of Medicine, University of Porto, Porto, Portugal.
| |
Collapse
|
22
|
Huan HB, Wen XD, Chen XJ, Wu L, Wu LL, Zhang L, Yang DP, Zhang X, Bie P, Qian C, Xia F. Sympathetic nervous system promotes hepatocarcinogenesis by modulating inflammation through activation of alpha1-adrenergic receptors of Kupffer cells. Brain Behav Immun 2017; 59:118-134. [PMID: 27585737 DOI: 10.1016/j.bbi.2016.08.016] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 08/23/2016] [Accepted: 08/27/2016] [Indexed: 02/07/2023] Open
Abstract
The sympathetic nervous system (SNS) is known to play a significant role in tumor initiation and metastasis. Hepatocellular carcinoma (HCC) frequently occurs in cirrhotic livers after chronic inflammation, and the SNS is hyperactive in advanced liver cirrhosis. However, it remains unclear whether the SNS promotes hepatocarcinogenesis by modulating chronic liver inflammation. In this study, a retrospective pathological analysis and quantification of sympathetic nerve fiber densities (tyrosine hydroxylase, TH+) in HCC patients, and diethylnitrosamine (DEN)-induced hepatocarcinogenesis in rats were performed. Our data showed that high density of sympathetic nerve fibers and α1-adrenergic receptors (ARs) of Kupffer cells (KCs) were associated with a poor prognosis of HCC. Sympathetic denervation or blocking of α1-ARs decreased DEN-induced HCC incidence and tumor development. In addition, synergistic effects of interleukin-6 (IL-6) and transforming growth factor-beta (TGF-β) in hepatocarcinogenesis were observed. The suppression of the SNS reduced IL-6 and TGF-β expression, which suppressed hepatocarcinogenesis, and KCs play a key role in this process. After the ablation of KCs, IL-6 and TGF-β expression and the development of HCC were inhibited. This study demonstrates that sympathetic innervation is crucial for hepatocarcinogenesis and that the SNS promotes hepatocarcinogenesis by activating α1-ARs of KCs to boost the activation of KCs and to maintain the inflammatory microenvironment. These results indicate that sympathetic denervation or α1-ARs blockage may represent novel treatment approaches for HCC.
Collapse
Affiliation(s)
- Hong-Bo Huan
- Institute of Hepatobiliary Surgery, Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Xu-Dong Wen
- Institute of Hepatobiliary Surgery, Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Xue-Jiao Chen
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, and Key Laboratory of Tumor Immunology and Pathology of Ministry of Education China, Chongqing, China
| | - Lin Wu
- Institute of Hepatobiliary Surgery, Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Li-Li Wu
- Institute of Hepatobiliary Surgery, Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Liang Zhang
- Institute of Hepatobiliary Surgery, Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Da-Peng Yang
- Institute of Hepatobiliary Surgery, Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Xia Zhang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, and Key Laboratory of Tumor Immunology and Pathology of Ministry of Education China, Chongqing, China
| | - Ping Bie
- Institute of Hepatobiliary Surgery, Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Cheng Qian
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, and Key Laboratory of Tumor Immunology and Pathology of Ministry of Education China, Chongqing, China
| | - Feng Xia
- Institute of Hepatobiliary Surgery, Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China.
| |
Collapse
|
23
|
The Role of α1-Adrenoceptor Antagonists in the Treatment of Prostate and Other Cancers. Int J Mol Sci 2016; 17:ijms17081339. [PMID: 27537875 PMCID: PMC5000736 DOI: 10.3390/ijms17081339] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 08/05/2016] [Accepted: 08/08/2016] [Indexed: 12/21/2022] Open
Abstract
This review evaluates the role of α-adrenoceptor antagonists as a potential treatment of prostate cancer (PCa). Cochrane, Google Scholar and Pubmed were accessed to retrieve sixty-two articles for analysis. In vitro studies demonstrate that doxazosin, prazosin and terazosin (quinazoline α-antagonists) induce apoptosis, decrease cell growth, and proliferation in PC-3, LNCaP and DU-145 cell lines. Similarly, the piperazine based naftopidil induced cell cycle arrest and death in LNCaP-E9 cell lines. In contrast, sulphonamide based tamsulosin did not exhibit these effects. In vivo data was consistent with in vitro findings as the quinazoline based α-antagonists prevented angiogenesis and decreased tumour mass in mice models of PCa. Mechanistically the cytotoxic and antitumor effects of the α-antagonists appear largely independent of α 1-blockade. The proposed targets include: VEGF, EGFR, HER2/Neu, caspase 8/3, topoisomerase 1 and other mitochondrial apoptotic inducing factors. These cytotoxic effects could not be evaluated in human studies as prospective trial data is lacking. However, retrospective studies show a decreased incidence of PCa in males exposed to α-antagonists. As human data evaluating the use of α-antagonists as treatments are lacking; well designed, prospective clinical trials are needed to conclusively demonstrate the anticancer properties of quinazoline based α-antagonists in PCa and other cancers.
Collapse
|
24
|
|
25
|
Tzirogiannis KN, Kourentzi KT, Zyga S, Papalimneou V, Tsironi M, Grypioti AD, Protopsaltis I, Panidis D, Panoutsopoulos GI. Effect of 5-HT7 receptor blockade on liver regeneration after 60-70% partial hepatectomy. BMC Gastroenterol 2014; 14:201. [PMID: 25433672 PMCID: PMC4267430 DOI: 10.1186/s12876-014-0201-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 11/11/2014] [Indexed: 01/05/2023] Open
Abstract
Background Serotonin exhibits a vast repertoire of actions including cell
proliferation and differentiation. The effect of serotonin, as an incomplete
mitogen, on liver regeneration has recently been unveiled and is mediated through
5-HT2 receptor. The aim of the present study was to
investigate the effect of 5-HT7 receptor blockade on liver
regeneration after partial hepatectomy. Methods Male Wistar rats were subjected to 60-70% partial hepatectomy.
5-HT7 receptor blockade was applied by intraperitoneal
administration of SB-269970 hydrochloride two hours prior to and sixteen hours
after partial hepatectomy and by intraperitoneal administration of SB-258719
sixteen hours after partial hepatectomy. Animals were sacrificed at different time
points until 72 h after partial hepatectomy. Liver regeneration was evaluated by
[3H]-thymidine incorporation into hepatic DNA, the
mitotic index in hematoxylin-eosin (HE) sections and by immunochemical detection
of Ki67 nuclear antigen. Reversion of 5-HT7 blockade was
performed by intraperitoneal administration of AS-19. Serum and liver tissue
lipids were also quantified. Results Liver regeneration peaked at 24 h
([3H]-thymidine incorporation into hepatic DNA and
mitotic index by immunochemical detection of Ki67) and at 32 h (mitotic index in
HE sections) in the control group of rats. 5-HT7 receptor
blockade had no effect on liver regeneration when applied 2 h prior to partial
hepatectomy. Liver regeneration was greatly attenuated when blockade of
5-HT7 receptor was applied (by SB-258719 and SB-269970)
at 16 h after partial hepatectomy and peaked at 32 h
([3H]-thymidine incorporation into hepatic DNA and
mitotic index by immunochemical detection of Ki67) and 40 h (mitotic index in HE
sections) after partial hepatectomy. AS-19 administration totally reversed the
observed attenuation of liver regeneration. Conclusions In conclusion, 5-HT7 receptor is a novel type
of serotonin receptor implicated in hepatocyte proliferation.
Collapse
Affiliation(s)
| | - Kalliopi T Kourentzi
- Department of Experimental Pharmacology, Medical School, Athens University, Athens, 11527, Greece.
| | - Sofia Zyga
- Department of Nursing, Faculty of Human Movement and Quality of Life Sciences, University of Peloponnese, Sparta, 23100, Greece.
| | | | - Maria Tsironi
- Department of Nursing, Faculty of Human Movement and Quality of Life Sciences, University of Peloponnese, Sparta, 23100, Greece.
| | - Agni D Grypioti
- Department of Experimental Pharmacology, Medical School, Athens University, Athens, 11527, Greece.
| | - Ioannis Protopsaltis
- Department of Internal Medicine, Tzanio General Hospital of Piraeus, Piraeus, 18537, Greece.
| | - Dimitrios Panidis
- Department of Nursing, Faculty of Human Movement and Quality of Life Sciences, University of Peloponnese, Sparta, 23100, Greece.
| | - Georgios I Panoutsopoulos
- Department of Nursing, Faculty of Human Movement and Quality of Life Sciences, University of Peloponnese, Orthias Artemidos and Plateon, Sparta, 23100, Greece.
| |
Collapse
|
26
|
Herrera-García AM, Domínguez-Luis MJ, Arce-Franco M, Armas-González E, Álvarez de La Rosa D, Machado JD, Pec MK, Feria M, Barreiro O, Sánchez-Madrid F, Díaz-González F. Prevention of neutrophil extravasation by α2-adrenoceptor-mediated endothelial stabilization. THE JOURNAL OF IMMUNOLOGY 2014; 193:3023-35. [PMID: 25114107 DOI: 10.4049/jimmunol.1400255] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Adrenergic receptors are expressed on the surface of inflammation-mediating cells, but their potential role in the regulation of the inflammatory response is still poorly understood. The objectives of this work were to study the effects of α2-adrenergic agonists on the inflammatory response in vivo and to determine their mechanism of action. In two mouse models of inflammation, zymosan air pouch and thioglycolate-induced peritonitis models, the i.m. treatment with xylazine or UK14304, two α2-adrenergic agonists, reduced neutrophil migration by 60%. The α2-adrenergic antagonist RX821002 abrogated this effect. In flow cytometry experiments, the basal surface expression of L-selectin and CD11b was modified neither in murine nor in human neutrophils upon α2-agonist treatment. Similar experiments in HUVEC showed that UK14304 prevented the activation-dependent upregulation of ICAM-1. In contrast, UK14304 augmented electrical resistance and reduced macromolecular transport through a confluent HUVEC monolayer. In flow chamber experiments, under postcapillary venule-like flow conditions, the pretreatment of HUVECs, but not neutrophils, with α2-agonists decreased transendothelial migration, without affecting neutrophil rolling. Interestingly, α2-agonists prevented the TNF-α-mediated decrease in expression of the adherens junctional molecules, VE-cadherin, β-catenin, and plakoglobin, and reduced the ICAM-1-mediated phosphorylation of VE-cadherin by immunofluorescence and confocal analysis and Western blot analysis, respectively. These findings indicate that α2-adrenoceptors trigger signals that protect the integrity of endothelial adherens junctions during the inflammatory response, thus pointing at the vascular endothelium as a therapeutic target for the management of inflammatory processes in humans.
Collapse
Affiliation(s)
- Ada María Herrera-García
- Departamento de Medicina, Facultad de Medicina, Universidad de La Laguna, La Cuesta, 38320 San Cristóbal de La Laguna, Santa Cruz de Tenerife, Spain
| | - María Jesús Domínguez-Luis
- Centro para la Investigación Biomédica de las Islas Canarias, Instituto de Tecnologías Biomedicas, Universidad de La Laguna, La Cuesta, 38320 San Cristóbal de La Laguna, Santa Cruz de Tenerife, Spain
| | - María Arce-Franco
- Servicio de Reumatología, Hospital Universitario de Canarias, La Cuesta, 38320 San Cristóbal de La Laguna, Santa Cruz de Tenerife, Spain
| | - Estefanía Armas-González
- Departamento de Medicina, Facultad de Medicina, Universidad de La Laguna, La Cuesta, 38320 San Cristóbal de La Laguna, Santa Cruz de Tenerife, Spain
| | - Diego Álvarez de La Rosa
- Departamento de Fisiología, Facultad de Medicina, Universidad de La Laguna, La Cuesta, 38320 San Cristóbal de La Laguna, Santa Cruz de Tenerife, Spain
| | - José David Machado
- Departamento de Farmacología, Facultad de Medicina, Universidad de La Laguna, La Cuesta, 38320 San Cristóbal de La Laguna, Santa Cruz de Tenerife, Spain
| | - Martina K Pec
- Departamento de Medicina, Facultad de Medicina, Universidad de La Laguna, La Cuesta, 38320 San Cristóbal de La Laguna, Santa Cruz de Tenerife, Spain
| | - Manuel Feria
- Departamento de Farmacología, Facultad de Medicina, Universidad de La Laguna, La Cuesta, 38320 San Cristóbal de La Laguna, Santa Cruz de Tenerife, Spain
| | - Olga Barreiro
- Departamento de Biología Vascular e Inflamación, Centro Nacional de Investigaciones Cardiovasculares, Consejo Superior de Investigaciones Cientificas, 28029 Madrid, Spain; and
| | - Francisco Sánchez-Madrid
- Departamento de Biología Vascular e Inflamación, Centro Nacional de Investigaciones Cardiovasculares, Consejo Superior de Investigaciones Cientificas, 28029 Madrid, Spain; and Servicio de Inmunología, Hospital Universitario de La Princesa, 28006 Madrid, Spain
| | - Federico Díaz-González
- Departamento de Medicina, Facultad de Medicina, Universidad de La Laguna, La Cuesta, 38320 San Cristóbal de La Laguna, Santa Cruz de Tenerife, Spain; Servicio de Reumatología, Hospital Universitario de Canarias, La Cuesta, 38320 San Cristóbal de La Laguna, Santa Cruz de Tenerife, Spain;
| |
Collapse
|
27
|
Jeon J, Nim S, Teyra J, Datti A, Wrana JL, Sidhu SS, Moffat J, Kim PM. A systematic approach to identify novel cancer drug targets using machine learning, inhibitor design and high-throughput screening. Genome Med 2014; 6:57. [PMID: 25165489 PMCID: PMC4143549 DOI: 10.1186/s13073-014-0057-7] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Accepted: 07/18/2014] [Indexed: 12/14/2022] Open
Abstract
We present an integrated approach that predicts and validates novel anti-cancer drug targets. We first built a classifier that integrates a variety of genomic and systematic datasets to prioritize drug targets specific for breast, pancreatic and ovarian cancer. We then devised strategies to inhibit these anti-cancer drug targets and selected a set of targets that are amenable to inhibition by small molecules, antibodies and synthetic peptides. We validated the predicted drug targets by showing strong anti-proliferative effects of both synthetic peptide and small molecule inhibitors against our predicted targets.
Collapse
Affiliation(s)
- Jouhyun Jeon
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1 Canada
| | - Satra Nim
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1 Canada
| | - Joan Teyra
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1 Canada
| | - Alessandro Datti
- Center for Systems Biology, Samuel Lunenfeld Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, ON M5S 3E1 Canada
- Department of Agricultural, Food and Environmental Sciences, University of Perugia, Perugia, 06100 Italy
| | - Jeffrey L Wrana
- Center for Systems Biology, Samuel Lunenfeld Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, ON M5S 3E1 Canada
| | - Sachdev S Sidhu
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1 Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 3E1 Canada
| | - Jason Moffat
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1 Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 3E1 Canada
| | - Philip M Kim
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S 3E1 Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 3E1 Canada
- Department of Computer Science, University of Toronto, Toronto, ON M5S 3E1 Canada
| |
Collapse
|
28
|
Wang L, Liu H, Chen X, Zhang M, Xie K, Ma Q. Immune sculpting of norepinephrine on MHC-I, B7-1, IDO and B7-H1 expression and regulation of proliferation and invasion in pancreatic carcinoma cells. PLoS One 2012; 7:e45491. [PMID: 23029049 PMCID: PMC3446877 DOI: 10.1371/journal.pone.0045491] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2011] [Accepted: 08/23/2012] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND The sympathetic neurotransmitter Norepinephrine (NE) contributes to tumorigenesis and cancer progression. This study aims to investigate the role of NE in modulating the immune phenotype and allowing pancreatic carcinoma (PC) cells to escape the immune response. METHODS Varied concentrations of NE and interferon-gamma (IFN-γ) were administrated to MIA PaCa-2 and BxPC-3 cell lines for 48 hours. Proliferation and invasion were then investigated using an MTT assay and a membrane invasion culture system respectively. MHC-I, B7-1, IDO and B7-H1 expression were measured using real-time quantitative RT-PCR, western blotting and immunocytochemistry. The synergistic and time-dependent effects of NE/IFN-γ were also investigated. Adrenergic antagonists were used to identify the relevant target receptor of NE. RESULTS The results showed that NE had dose-dependent and time-dependent effects on cell biological processes as well as on the expression of MHC-I, B7-1, IDO and B7-H1. These effects occurred mainly via the β(2)-adrenergic receptor. Long-term NE treatment was able to antagonize some of the effects of IFN-γ (after 2 weeks of treatment), but NE and IFN-γ had significant synergistic stimulatory effects on IDO and B7-H1 expression. The residual effects on biological activities lasted for 2 weeks, while the immunophenotypic changes decreased at early time points after treatment. CONCLUSIONS NE plays important roles in modulating PC cell biological activities and affecting MHC-I, B7-1, IDO and B7-H1 expression in vitro, mainly via the β2-adrenergic receptor (β2-AR) in a time- and dose-dependent fashion. Only at extended treatment durations could NE affect PC cell progression and immune evasion.
Collapse
Affiliation(s)
- Liancai Wang
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi Province, China
- Henan Province People’s Hospital, Zhengzhou, Henan Province, China
| | - Han Liu
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi Province, China
| | - Xiangli Chen
- Henan Province People’s Hospital, Zhengzhou, Henan Province, China
| | - Min Zhang
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi Province, China
| | - Keping Xie
- Department of Gastrointestinal Medical Oncology, the University of Texas, MD Anderson Cancer Centre, Houston, Texas
| | - Qingyong Ma
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi Province, China
| |
Collapse
|
29
|
Pérez-Sayáns M, Somoza-Martín JM, Barros-Angueira F, Gayoso-Diz P, Otero-Rey EM, Gándra-Rey JM, García-García A. Activity of β2-adrenergic receptor in oral squamous cell carcinoma is mediated by overexpression of the ADRBK2 gene: a pilot study. Biotech Histochem 2011; 87:179-86. [PMID: 21916780 DOI: 10.3109/10520295.2011.592151] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The β2-adrenergic receptor is most frequently involved in carcinogenic processes. Earlier studies have established a relation between the β2-adrenergic receptor and various characteristics of cancer including cell proliferation, apoptosis, chemotaxis, metastasis, tumor growth and angiogenesis. Our goal was to determine differential expression of the genes involved in adrenergic receptors using DNA microarrays and to confirm their under- or overexpression using real-time quantitative PCR. Five of the nine genes investigated showed significantly altered expression levels in tumor cells (p < 0.05). The gene product with the highest Z-score (restrictive statistical technique for selection of appropriate genes to study) was ADRBK2. Significantly, most of the overexpressed genes were related to β-adrenergic receptors. Real-time PCR analysis confirmed the up regulation observed in the microarrays, which indicated overexpression in 100% of the tumors. In oral squamous cell carcinomas, malignant cells and surrounding tissue overexpress the ADRBK2 gene.
Collapse
Affiliation(s)
- M Pérez-Sayáns
- Oral Medicine, Oral Surgery and Implantology Unit, Faculty of Medicine and Dentistry, Entrerríos s/n, Santiago de Compostela, Spain.
| | | | | | | | | | | | | |
Collapse
|
30
|
Raju B, Ibrahim SO. Pathophysiology of oral cancer in experimental animal models: a review with focus on the role of sympathetic nerves. J Oral Pathol Med 2010; 40:1-9. [PMID: 20819130 DOI: 10.1111/j.1600-0714.2010.00928.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Global increase in incidence and mortality as well as poor prognosis of oral cancer (OC) has intensified efforts towards early detection and prevention of this disfiguring disease. Several studies have been conducted using experimental animal models to understand the pathophysiology and molecular events involved in OC. Lack of identification of specific biomarkers during the multifaceted steps of oral carcinogenesis has hindered its diagnosis and treatment. Solid stress generated by growing tumors as well as abnormalities in tumor vasculature lead to increased interstitial fluid pressure, which could obstruct therapeutic drug delivery to tumors. Furthermore, the sympathetic nervous system is known to affect angiogenesis, vessel permeability, immune responses and carcinogenesis. Recent findings indicate that, in addition to angiogenic and lymphangiogenic factors, tumor cells release neurotrophic factors that initiate innervation. Interactions between cytokines and sympathetic neurotransmitters, and their respective receptors expressed by the nerve, immune and tumor cells appear to influence tumor growth. Thus, understanding the complex signaling processes and interrelationships between vascular, nervous and immune systems during oral carcinogenesis may prove vital for successful prevention and treatment of OC. This review aims at outlining the available knowledge on pathophysiology of OC in experimental animal models including evidence from our own findings.
Collapse
Affiliation(s)
- Bina Raju
- Department of Biomedicine, Section for Biochemistry and Molecular Biology, University of Bergen, Bergen, Norway
| | | |
Collapse
|
31
|
Oben JA, Patel T, Mouralidarane A, Samuelsson AM, Matthews P, Pombo J, Morgan M, McKee C, Soeda J, Novelli M, Poston L, Taylor P. Maternal obesity programmes offspring development of non-alcoholic fatty pancreas disease. Biochem Biophys Res Commun 2010; 394:24-8. [PMID: 20170634 PMCID: PMC2877817 DOI: 10.1016/j.bbrc.2010.02.057] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2010] [Accepted: 02/11/2010] [Indexed: 01/09/2023]
Abstract
BACKGROUND AND AIMS The prevalence of pancreatic adenocarcinoma (PAC) parallels rising rates of obesity and dysmetabolism, a possible link being non-alcoholic fatty pancreas disease (NAFPD). We have recently shown that maternal obesity programmes the development of a dysmetabolic and fatty liver (non-alcoholic fatty liver disease, NAFLD) phenotype in adult offspring. Since the pancreas and liver originate from the same embryonic bud, it is plausible that maternal obesity may similarly programme the development of NAFPD. Our objective was to determine the effect of maternal obesity on development of NAFPD in offspring and ascertain contributions of the intra/extra-uterine periods. METHODS Female C57BL/6J mice were fed either a standard chow (3% fat, 7% sugar) or a hypercalorific diet (16% fat, 33% sugar) for six weeks prior to mating and throughout pregnancy and lactation. Female offspring were cross-fostered for suckling to dams on the same or opposite diet to yield four groups: offspring of lean suckled by lean dams (n=6), offspring of obese suckled by obese dams (n=6), offspring of lean suckled by obese dams (n=5) and offspring of obese suckled by lean dams (n=6). All offspring were weaned onto a standard chow diet at 21 days and sacrificed at 3 months post-partum for tissue collection. RESULTS Offspring subjected to an adverse suckling environment showed significant increases in body weight, pancreatic triglyceride content, TGF-beta, collagen gene expression and SBP at rest along with an enhanced restraint stress response, indicating a dysmetabolic and NAFPD phenotype. CONCLUSIONS Developmental programming is involved in the pathogenesis of NAFPD and appears to be largely dependent on an adverse extra-uterine environment.
Collapse
Affiliation(s)
- Jude A Oben
- Centre for Hepatology, University College London, Royal Free Hospital, London NW3 2PF, UK.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
HIF-1alpha links beta-adrenoceptor agonists and pancreatic cancer cells under normoxic condition. Acta Pharmacol Sin 2010; 31:102-10. [PMID: 20037603 DOI: 10.1038/aps.2009.181] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
AIM To examine whether beta-adrenoceptor (beta-AR) agonists can induce hypoxia-inducible factor (HIF)-1alpha accumulation which then up-regulate the expression of its target genes in pancreatic cancer cells at normoxia, and to further elucidate the mechanism involved. METHODS Pulse-chase assay, RT-PCR, and Western blot were employed to detect the effects of beta-AR agonists and antagonists, siRNA as well as several inhibitors of signal transduction pathways on MIA PaCa2 and BxPC-3 pancreatic cancer cells. RESULTS Treatment of pancreatic cancer cell lines with beta-AR agonists led to accumulation of HIF-1alpha and then up-regulated expression of its target genes independently of oxygen levels. The induction was partly or completely inhibited not only by beta-AR antagonists but also by inhibitors of PKA transduction pathways and by siHIF-1alpha. Both beta1-AR and beta2-AR agonists produced the above-mentioned effects, but beta2-AR agonist was more potent. CONCLUSION Activation of beta-AR receptor transactivates epidermal growth factor receptor (EGFR) and then elicits Akt and ERK1/2 in a PKA-dependent manner, which together up-regulate levels of HIF-1alpha and downstream target genes independently of oxygen level. Our data suggest a novel mechanism in pancreatic cancer cells that links beta-AR and HIF-1alpha signaling under normoxic conditions, with implications for the control of glucose transport, angiogenesis and metastasis.
Collapse
|