1
|
Dong ZK, Wang YF, Li WP, Jin WL. Neurobiology of cancer: Adrenergic signaling and drug repurposing. Pharmacol Ther 2024; 264:108750. [PMID: 39527999 DOI: 10.1016/j.pharmthera.2024.108750] [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: 07/04/2024] [Revised: 10/04/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Cancer neuroscience, as an emerging converging discipline, provides us with new perspectives on the interactions between the nervous system and cancer progression. As the sympathetic nervous system, in particular adrenergic signaling, plays an important role in the regulation of tumor activity at every hierarchical level of life, from the tumor cell to the tumor microenvironment, and to the tumor macroenvironment, it is highly desirable to dissect its effects. Considering the far-reaching implications of drug repurposing for antitumor drug development, such a large number of adrenergic receptor antagonists on the market has great potential as one of the means of antitumor therapy, either as primary or adjuvant therapy. Therefore, this review aims to summarize the impact of adrenergic signaling on cancer development and to assess the status and prospects of intervening in adrenergic signaling as a therapeutic tool against tumors.
Collapse
Affiliation(s)
- Zi-Kai Dong
- The First Clinical Medical College of Lanzhou University, Lanzhou 730000, PR China; Institute of Cancer Neuroscience, Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, The First Clinical Medical College of Lanzhou University, Lanzhou 730000, PR China
| | - Yong-Fei Wang
- The First Clinical Medical College of Lanzhou University, Lanzhou 730000, PR China; Institute of Cancer Neuroscience, Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, The First Clinical Medical College of Lanzhou University, Lanzhou 730000, PR China
| | - Wei-Ping Li
- The First Clinical Medical College of Lanzhou University, Lanzhou 730000, PR China; Department of Urology, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, PR China
| | - Wei-Lin Jin
- The First Clinical Medical College of Lanzhou University, Lanzhou 730000, PR China; Institute of Cancer Neuroscience, Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, The First Clinical Medical College of Lanzhou University, Lanzhou 730000, PR China.
| |
Collapse
|
2
|
Zhang H, Yang Y, Cao Y, Guan J. Effects of chronic stress on cancer development and the therapeutic prospects of adrenergic signaling regulation. Biomed Pharmacother 2024; 175:116609. [PMID: 38678960 DOI: 10.1016/j.biopha.2024.116609] [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: 01/19/2024] [Revised: 04/14/2024] [Accepted: 04/17/2024] [Indexed: 05/01/2024] Open
Abstract
Long-term chronic stress is an important factor in the poor prognosis of cancer patients. Chronic stress reduces the tissue infiltration of immune cells in the tumor microenvironment (TME) by continuously activating the adrenergic signaling, inhibits antitumor immune response and tumor cell apoptosis while also inducing epithelial-mesenchymal transition (EMT) and tumor angiogenesis, promoting tumor invasion and metastasis. This review first summarizes how adrenergic signaling activates intracellular signaling by binding different adrenergic receptor (AR) heterodimers. Then, we focused on reviewing adrenergic signaling to regulate multiple functions of immune cells, including cell differentiation, migration, and cytokine secretion. In addition, the article discusses the mechanisms by which adrenergic signaling exerts pro-tumorigenic effects by acting directly on the tumor itself. It also highlights the use of adrenergic receptor modulators in cancer therapy, with particular emphasis on their potential role in immunotherapy. Finally, the article reviews the beneficial effects of stress intervention measures on cancer treatment. We think that enhancing the body's antitumor response by adjusting adrenergic signaling can enhance the efficacy of cancer treatment.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Oncology, The Eighth Medical Center, Chinese PLA (People's Liberation Army) General Hospital, Beijing 100091, China; Department of Oncology, The Fifth Medical Center, Chinese PLA (People's Liberation Army) General Hospital, Beijing 100071, China.
| | - Yuwei Yang
- College of Pulmonary & Critical Care Medicine, Chinese PLA General Hospital, Beijing Key Laboratory of OTIR, Beijing, 100091, China.
| | - Yan Cao
- College of Pulmonary & Critical Care Medicine, Chinese PLA General Hospital, Beijing Key Laboratory of OTIR, Beijing, 100091, China.
| | - Jingzhi Guan
- Department of Oncology, The Fifth Medical Center, Chinese PLA (People's Liberation Army) General Hospital, Beijing 100071, China.
| |
Collapse
|
3
|
Pasha A, Tondo A, Favre C, Calvani M. Inside the Biology of the β3-Adrenoceptor. Biomolecules 2024; 14:159. [PMID: 38397396 PMCID: PMC10887351 DOI: 10.3390/biom14020159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 01/24/2024] [Accepted: 01/27/2024] [Indexed: 02/25/2024] Open
Abstract
Since the first discovery in 1989, the β3-adrenoceptor (β3-AR) has gained great attention because it showed the ability to regulate many physiologic and metabolic activities, such as thermogenesis and lipolysis in brown and white adipose tissue, respectively (BAT, WAT), negative inotropic effects in cardiomyocytes, and relaxation of the blood vessels and the urinary bladder. The β3-AR has been suggested as a potential target for cancer treatment, both in adult and pediatric tumors, since under hypoxia its upregulation in the tumor microenvironment (TME) regulates stromal cell differentiation, tumor growth and metastases, signifying that its agonism/antagonism could be useful for clinical benefits. Promising results in cancer research have proposed the β3-AR being targeted for the treatment of many conditions, with some drugs, at present, undergoing phase II and III clinical trials. In this review, we report the scientific journey followed by the research from the β3-Ars' discovery, with focus on the β3-Ars' role in cancer initiation and progression that elects it an intriguing target for novel antineoplastic approaches. The overview highlights the great potential of the β3-AR, both in physiologic and pathologic conditions, with the intention to display the possible benefits of β3-AR modulation in cancer reality.
Collapse
Affiliation(s)
- Amada Pasha
- Department of Pediatric Hematology–Oncology, Meyer Children’s Hospital IRCCS, 50139 Florence, Italy; (A.P.); (A.T.); (C.F.)
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50139 Florence, Italy
| | - Annalisa Tondo
- Department of Pediatric Hematology–Oncology, Meyer Children’s Hospital IRCCS, 50139 Florence, Italy; (A.P.); (A.T.); (C.F.)
| | - Claudio Favre
- Department of Pediatric Hematology–Oncology, Meyer Children’s Hospital IRCCS, 50139 Florence, Italy; (A.P.); (A.T.); (C.F.)
| | - Maura Calvani
- Department of Pediatric Hematology–Oncology, Meyer Children’s Hospital IRCCS, 50139 Florence, Italy; (A.P.); (A.T.); (C.F.)
| |
Collapse
|
4
|
Jabloñski M, Rodríguez MS, Rivero EM, Bruque CD, Vanzulli S, Bruzzone A, Pérez Piñero C, Lüthy IA. The Beta2-adrenergic agonist salbutamol synergizes with paclitaxel on cell proliferation and tumor growth in triple negative breast cancer models. Cancer Chemother Pharmacol 2023; 92:485-499. [PMID: 37725114 DOI: 10.1007/s00280-023-04586-9] [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: 03/27/2023] [Accepted: 08/31/2023] [Indexed: 09/21/2023]
Abstract
PURPOSE Globally breast cancer accounts for 24.5% in incidence and 15.5% in cancer deaths in women. The triple-negative subtype lacks any specific therapy and is treated with chemotherapy, resulting in significant side-effects. We aimed to investigate if the dose of chemotherapeutic drugs could be diminished by co-administering it with the β2-agonist salbutamol. METHODS Cell proliferation was measured by thymidine incorporation; gene expression, by real-time PCR and protein phosphorylation by WB. Apoptosis was assessed by acridine orange / ethidium bromide and TUNEL tests. Public patient databases were consulted. Cells were inoculated to nude mice and their growth assessed. RESULTS The β2-agonist salbutamol synergizes in MDA-MB-231 cells in vitro with paclitaxel and doxorubicin on cell proliferation through ADRB2 receptors, while the β-blocker propranolol does not. The expression of this receptor was assessed in patient databases and other cell lines. Triple negative samples had the lowest expression. Salbutamol and paclitaxel decreased MDA-MB-231 cell proliferation while their combination further inhibited it. The pathways involved were analyzed. When these cells were inoculated to nude mice, paclitaxel and salbutamol inhibited tumor growth. The combined effect was significantly greater. Paclitaxel increased the expression of MDR1 while salbutamol partially reversed this increase. CONCLUSION While the effect of salbutamol was mainly on cell proliferation, suboptimal concentrations of paclitaxel provoked a very important enhancement of apoptosis. The latter enhanced transporter proteins as MDR1, whose expression were diminished by salbutamol. The expression of ADRB2 should be assessed in the biopsy or tumor to eventually select patients that could benefit from salbutamol repurposing.
Collapse
Affiliation(s)
- Martina Jabloñski
- Instituto de Biología y Medicina Experimental (IBYME-CONICET), Obligado 2490, Ciudad Autónoma de Buenos Aires, Argentina
| | - María Sol Rodríguez
- Instituto de Biología y Medicina Experimental (IBYME-CONICET), Obligado 2490, Ciudad Autónoma de Buenos Aires, Argentina
| | - Ezequiel Mariano Rivero
- Instituto de Biología y Medicina Experimental (IBYME-CONICET), Obligado 2490, Ciudad Autónoma de Buenos Aires, Argentina
- Centre for Genomic Regulation, Barcelona, Spain
| | - Carlos David Bruque
- Unidad de Conocimiento Traslacional Hospitalaria Patagónica, Hospital de Alta Complejidad SAMIC - El Calafate, El Calafate, Argentina
| | | | - Ariana Bruzzone
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB-CONICET), Bahía Blanca, Argentina
| | - Cecilia Pérez Piñero
- Instituto de Biología y Medicina Experimental (IBYME-CONICET), Obligado 2490, Ciudad Autónoma de Buenos Aires, Argentina.
| | - Isabel Alicia Lüthy
- Instituto de Biología y Medicina Experimental (IBYME-CONICET), Obligado 2490, Ciudad Autónoma de Buenos Aires, Argentina.
| |
Collapse
|
5
|
Zidan A, El Saadany AA, El Maghraby GM, Abdin AA, Hedya SE. Potential cardioprotective and anticancer effects of carvedilol either free or as loaded nanoparticles with or without doxorubicin in solid Ehrlich carcinoma-bearing mice. Toxicol Appl Pharmacol 2023; 465:116448. [PMID: 36921847 DOI: 10.1016/j.taap.2023.116448] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 02/17/2023] [Accepted: 03/02/2023] [Indexed: 03/14/2023]
Abstract
AIM The aim of this study was to investigate the potential cardioprotective and anti-cancer effects of carvedilol (CAR) either free or as loaded nano-formulated with or without doxorubicin (DOX) in solid Ehrlich carcinoma (SEC)-bearing mice. It focused on assessment of cardiac damage, drug resistance, apoptosis, oxidative stress status, angiogenesis and proliferation. METHODS CAR was loaded into poly-D,L lactic-co-glycolic acid)PLGA(or Niosomes. SEC was induced in female albino mice as an experimental model of breast cancer. Seventy-two mice were randomly divided into 9 equal groups (Normal control, Untreated-SEC, SEC + DOX, SEC + CAR-free, SEC + CAR-PLGA, SEC + CAR-Niosomes, SEC + DOX + CAR-free, SEC + DOX + CAR-PLGA and SEC + DOX + CAR-Niosomes). Tumor volume and survival rate were recorded. On day 28 from tumor inoculation, mice were sacrificed, and blood samples were collected for determination of serum lactate dehydrogenase (LDH) and creatine kinase-MB (CK-MB). One part from tumor tissues was prepared for assessment of multidrug resistance protein-1 (MDR-1), caspase-3, reduced glutathione (GSH) and malondialdehyde (MDA), while the other part was processed for histopathological examination and immunohistochemical expression of vascular endothelial growth factor (VEGF) and Ki-67. RESULTS There was non-significant difference between CAR-free, CAR-PLGA and CAR-Niosomes as anticancer either alone or when combined with DOX. However, CAR-free demonstrated potential cardioprotective effects against cardiac damage mediated by cancer or DOX that have been enhanced using CAR-PLGA or CAR-Niosomes, but that of Niosomes outperformed them both. CONCLUSION CAR could be used as an adjuvant therapy with DOX, especially when nanoformualted with PLGA and even better with Niosomes, without compromising its cytotoxicity against cancer cells and preventing its cardiotoxic impacts.
Collapse
Affiliation(s)
- Amr Zidan
- Department of Pharmacology, Faculty of Medicine, Tanta University, Egypt.
| | - Amira A El Saadany
- Department of Pharmacology, Faculty of Medicine, Tanta University, Egypt
| | - Gamal M El Maghraby
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Tanta University, Egypt
| | - Amany A Abdin
- Department of Pharmacology, Faculty of Medicine, Tanta University, Egypt
| | - Sabeha E Hedya
- Department of Pharmacology, Faculty of Medicine, Tanta University, Egypt
| |
Collapse
|
6
|
Ji L, Xu F, Zhang J, Song T, Chen W, Yin X, Wang Q, Chen X, Li X, Guo M, Chen Z. ADRB2 expression predicts the clinical outcomes and is associated with immune cells infiltration in lung adenocarcinoma. Sci Rep 2022; 12:15994. [PMID: 36163241 PMCID: PMC9512930 DOI: 10.1038/s41598-022-19991-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 09/07/2022] [Indexed: 11/09/2022] Open
Abstract
The gene encoding beta2-adrenergic receptor (β2-AR), adrenoceptor beta 2 (ADRB2), has been reported to closely associated with various cancers. However, its role in lung adenocarcinoma (LUAD) remains controversial. This research shed light on the prognostic value of ADRB2 in LUAD and further explored its association with immune cell infiltration. ADRB2 was significantly decreased in LUAD. ADRB2 expression in LUAD was significantly correlated with gender, smoking status, T classification, and pathologic stage. Patients in the low ADRB2 expression group presented with significantly poorer overall survival (OS) and disease-specific survival (DSS). Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Set Enrichment Analysis (GSEA) results showed that ADRB2 participates in immune response. The expression of ADRB2 was positively correlated with the infiltration level of most immune cells. Notably, ADRB2 is involved in LUAD progression partly by regulating the immune microenvironment, which may potentially serve as a significant prognostic biomarker as well as a potential drug target.
Collapse
Affiliation(s)
- Lingyun Ji
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Fei Xu
- Department of Geriatric Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jingtao Zhang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ting Song
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Weida Chen
- Department of Geriatric Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xi Yin
- Department of Geriatric Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Qingqing Wang
- Department of Record Room, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, China, Jinan
| | - Xiubao Chen
- Department of Geriatric Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xin Li
- Department of Geriatric Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Minghao Guo
- Department of Geriatric Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zetao Chen
- Department of Geriatric Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China. .,Subject of Integrated Chinese and Western Medicine , Shandong University of Traditional Chinese Medicine, Jinan, China.
| |
Collapse
|
7
|
Pérez Piñero C, Rivero EM, Gargiulo L, Rodríguez MS, Bruque CD, Bruzzone A, Lüthy IA. Adrenergic receptors in breast cancer. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2022; 193:37-63. [PMID: 36357079 DOI: 10.1016/bs.pmbts.2022.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Breast cancer is the most diagnosed malignancy in women worldwide and in the majority of the countries. Breast cancers are classified on the expression of estrogen and progesterone receptor expression and overexpression of human epidermal growth factor receptor 2 (HER2) as luminal, HER2+ and triple negative breast cancer. The intrinsic molecular subtypes match this classification. Cancer diagnosis and treatment cause distress. In both acute and chronic stress, the secreted catecholamines adrenaline and noradrenaline trigger the "fight-or-flight" response. This chapter focuses on the actions of the β2 and α2 adrenergic receptors in several models of breast cancer. The actions of these receptors depend on the model used to investigate them. The β2-adrenergic receptors seem to exert a dual action. They can directly act on the epithelial cells inhibiting cell proliferation and migration/invasion and indirectly upon the immune microenvironment. The proportion of β2 receptors in each compartment could, therefore, lean the scale to an inhibition or to an exacerbation of tumor growth, invasion and metastasis. All the work points to a beneficial or neutral action of β-blockers on breast cancer. With respect to α2-adrenergic receptors, the investigation performed by our group suggest that the α2B and the α2C receptors are linked to enhanced cell proliferation and tumor growth acting through both the epithelial and the stromal (fibroblastic) compartments while α2A could be beneficial for patients. Some adrenergic compounds could be repurposed for breast cancer treatment due to their very low side effects and very well-known pharmacology.
Collapse
Affiliation(s)
- Cecilia Pérez Piñero
- Instituto de Biología y Medicina Experimental, IBYME-CONICET, Buenos Aires, Argentina
| | | | - Lucía Gargiulo
- Instituto de Biología y Medicina Experimental, IBYME-CONICET, Buenos Aires, Argentina
| | - María Sol Rodríguez
- Instituto de Biología y Medicina Experimental, IBYME-CONICET, Buenos Aires, Argentina
| | - Carlos David Bruque
- Genética Molecular Humana y Bioinformática, Unidad de Conocimiento Traslacional Hospitalaria Patagónica, Hospital de Alta Complejidad SAMIC - El Calafate, El Calafate, Argentina
| | - Ariana Bruzzone
- Instituto de Investigaciones Bioquímicas Bahía Blanca INIBIBB -CONICET, Buenos Aires, Argentina
| | - Isabel Alicia Lüthy
- Instituto de Biología y Medicina Experimental, IBYME-CONICET, Buenos Aires, Argentina.
| |
Collapse
|
8
|
Silva D, Quintas C, Gonçalves J, Fresco P. Contribution of adrenergic mechanisms for the stress-induced breast cancer carcinogenesis. J Cell Physiol 2022; 237:2107-2127. [PMID: 35243626 DOI: 10.1002/jcp.30707] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/21/2022] [Accepted: 02/12/2022] [Indexed: 12/11/2022]
Abstract
Breast cancer is the most common and deadliest type of cancer in women. Stress exposure has been associated with carcinogenesis and the stress released neurotransmitters, noradrenaline and adrenaline, and their cognate receptors, can participate in the carcinogenesis process, either by regulating tumor microenvironment or by promoting systemic changes. This work intends to provide an overview of the research done in this area and try to unravel the role of adrenergic ligands in the context of breast carcinogenesis. In the initiation phase, adrenergic signaling may favor neoplastic transformation of breast epithelial cells whereas, during cancer progression, may favor the metastatic potential of breast cancer cells. Additionally, adrenergic signaling can alter the function and activity of other cells present in the tumor microenvironment towards a protumor phenotype, namely macrophages, fibroblasts, and by altering adipocyte's function. Adrenergic signaling also promotes angiogenesis and lymphangiogenesis and, systemically, may induce the formation of preneoplastic niches, cancer-associated cachexia and alterations in the immune system which contribute for the loss of quality of life of breast cancer patients and their capacity to fight cancer. Most studies points to a major contribution of β2 -adrenoceptor activated pathways on these effects. The current knowledge of the mechanistic pathways activated by β2 -adrenoceptors in physiology and pathophysiology, the availability of selective drugs approved for clinical use and a deeper knowledge of the basic cellular and molecular pathways by which adrenergic stimulation may influence cancer initiation and progression, opens the possibility to use new therapeutic alternatives to improve efficacy of breast cancer treatments.
Collapse
Affiliation(s)
- Dany Silva
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Clara Quintas
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Jorge Gonçalves
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Paula Fresco
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| |
Collapse
|
9
|
Chaudhary B, Kumar P, Arya P, Singla D, Kumar V, Kumar D, S R, Wadhwa S, Gulati M, Singh SK, Dua K, Gupta G, Gupta MM. Recent Developments in the Study of the Microenvironment of Cancer and Drug Delivery. Curr Drug Metab 2022; 23:1027-1053. [PMID: 36627789 DOI: 10.2174/1389200224666230110145513] [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: 06/16/2022] [Revised: 09/20/2022] [Accepted: 11/29/2022] [Indexed: 01/12/2023]
Abstract
Cancer is characterized by disrupted molecular variables caused by cells that deviate from regular signal transduction. The uncontrolled segment of such cancerous cells annihilates most of the tissues that contact them. Gene therapy, immunotherapy, and nanotechnology advancements have resulted in novel strategies for anticancer drug delivery. Furthermore, diverse dispersion of nanoparticles in normal stroma cells adversely affects the healthy cells and disrupts the crosstalk of tumour stroma. It can contribute to cancer cell progression inhibition and, conversely, to acquired resistance, enabling cancer cell metastasis and proliferation. The tumour's microenvironment is critical in controlling the dispersion and physiological activities of nano-chemotherapeutics which is one of the targeted drug therapy. As it is one of the methods of treating cancer that involves the use of medications or other substances to specifically target and kill off certain subsets of malignant cells. A targeted therapy may be administered alone or in addition to more conventional methods of care like surgery, chemotherapy, or radiation treatment. The tumour microenvironment, stromatogenesis, barriers and advancement in the drug delivery system across tumour tissue are summarised in this review.
Collapse
Affiliation(s)
- Benu Chaudhary
- Department of Pharmacology, Guru Gobind Singh College of Pharmacy, Yamunanagar, Haryana, India
| | - Parveen Kumar
- Department of Life Science, Shri Ram College of Pharmacy, Karnal, Haryana, India
| | - Preeti Arya
- Department of Pharmacology, Guru Gobind Singh College of Pharmacy, Yamunanagar, Haryana, India
| | - Deepak Singla
- Department of Pharmacology, Guru Gobind Singh College of Pharmacy, Yamunanagar, Haryana, India
| | - Virender Kumar
- Department of Pharmacology, Swami Dayanand Post Graduate Institute of Pharmaceutical Sciences, Rohtak, Haryana, India
| | - Davinder Kumar
- Department of Pharmacology, Swami Dayanand Post Graduate Institute of Pharmaceutical Sciences, Rohtak, Haryana, India
| | - Roshan S
- Department of Pharmacology, Deccan School of Pharmacy, Hyderabad, India
| | - Sheetu Wadhwa
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - Monica Gulati
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, 144411, India
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, NSW 2007, Australia
| | - Gaurav Gupta
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Mahal Road, Jaipur, India
- Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Madan Mohan Gupta
- Faculty of Medical Sciences, School of Pharmacy, The University of the West Indies, St. Augustine, Trinidad & Tobago, West Indies
| |
Collapse
|
10
|
Lu X, Ma P, Kong L, Wang X, Wang Y, Jiang L. Vitamin K2 Inhibits Hepatocellular Carcinoma Cell Proliferation by Binding to 17β-Hydroxysteroid Dehydrogenase 4. Front Oncol 2021; 11:757603. [PMID: 34858832 PMCID: PMC8630649 DOI: 10.3389/fonc.2021.757603] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 10/18/2021] [Indexed: 12/31/2022] Open
Abstract
Our previous studies have proved that 17β-hydroxysteroid dehydrogenase 4 (HSD17B4) is a novel proliferation-promoting protein. The overexpression of HSD17B4 promotes hepatocellular carcinoma (HCC) cell proliferation. Vitamin K2 (VK2), a fat-soluble vitamin, has the function of promoting coagulation and can inhibit the progression of liver cancer. A previous study demonstrated that VK2 could bind to HSD17B4 in HepG2 cells. However, the mechanism of VK2 in inhibiting HCC cell proliferation is not clear. In this study, we investigate whether VK2 can inhibit the proliferation of HCC cell induced by HSD17B4 and the possible mechanism. We detected the effect of VK2 on HSD17B4-induced HCC cell proliferation, and the activation of STAT3, AKT, and MEK/ERK signaling pathways. We measured the effect of HSD17B4 on the growth of transplanted tumor and the inhibitory effect of VK2. Our results indicated that VK2 directly binds to HSD17B4, but does not affect the expression of HSD17B4, to inhibit the proliferation of HCC cells by inhibiting the activation of Akt and MEK/ERK signaling pathways, leading to decreased STAT3 activation. VK2 also inhibited the growth of HSD17B4-induced transplanted tumors. These findings provide a theoretical and experimental basis for possible future prevention and treatment of HCC using VK2.
Collapse
Affiliation(s)
- Xin Lu
- Center for Prenatal Diagnosis and Genetic Diseases, Tangshan Maternal and Children Hospital, Tangshan, China
| | - Panpan Ma
- Department of Blood Transfusion, Hebei General Hospital, Shijiazhuang, China
| | - Lingyu Kong
- Department of Oncology, Tianjin Binhai New Area Hospital of Traditional Chinese Medicine, Tianjin, China
| | - Xi Wang
- Department of Endocrinology, The Third Affiliated Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yaqi Wang
- Department of Clinical Laboratory, Hebei Provincial Hospital of Traditional Chinese Medicine, Shijiazhuang, China
| | - Lingling Jiang
- Department of Biochemistry and Molecular Biology, The Key Laboratory of Neurobiology and Vascular Biology, Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
11
|
Dabaghi M, Rasa SMM, Cirri E, Ori A, Neri F, Quaas R, Hilger I. Iron Oxide Nanoparticles Carrying 5-Fluorouracil in Combination with Magnetic Hyperthermia Induce Thrombogenic Collagen Fibers, Cellular Stress, and Immune Responses in Heterotopic Human Colon Cancer in Mice. Pharmaceutics 2021; 13:pharmaceutics13101625. [PMID: 34683917 PMCID: PMC8541380 DOI: 10.3390/pharmaceutics13101625] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/29/2021] [Accepted: 09/30/2021] [Indexed: 01/01/2023] Open
Abstract
In this study we looked for the main protein pathway regulators which were responsible for the therapeutic impact on colon cancers when combining magnetic hyperthermia with the chemotherapeutic agent 5-fluorouracil (5FU). To this end, chitosan-coated magnetic nanoparticles (MNP) functionalized with 5FU were intratumorally injected into subcutaneous human colon cancer xenografts (HT-29) in mice and exposed to an alternating magnetic field. A decreased tumor growth was found particularly for the combined thermo-chemotherapy vs. the corresponding monotherapies. By using computational analysis of the tumor proteome, we found upregulated functional pathway categories termed "cellular stress and injury", "intracellular second messenger and nuclear receptor signaling", "immune responses", and "growth proliferation and development". We predict TGF-beta, and other mediators, as important upstream regulators. In conclusion, our findings show that the combined thermo-chemotherapy induces thrombogenic collagen fibers which are able to impair tumor nutrient supply. Further on, we associate several responses to the recognition of damage associated molecular patterns (DAMPs) by phagocytic cells, which immigrate into the tumor area. The activation of some pathways associated with cell survival implies the necessity to conduct multiple therapy sessions in connection with a corresponding monitoring, which could possibly be performed on the base of the identified protein regulators.
Collapse
Affiliation(s)
- Mohammad Dabaghi
- Institute of Diagnostic and Interventional Radiology, Jena University Hospital, Friedrich Schiller University Jena, Am Klinikum 1, D-07740 Jena, Germany;
| | - Seyed Mohammad Mahdi Rasa
- Leibniz Institute on Aging Fritz Lipmann Institute (FLI), Beutenbergstraße 11, 07745 Jena, Germany; (S.M.M.R.); (E.C.); (A.O.); (F.N.)
| | - Emilio Cirri
- Leibniz Institute on Aging Fritz Lipmann Institute (FLI), Beutenbergstraße 11, 07745 Jena, Germany; (S.M.M.R.); (E.C.); (A.O.); (F.N.)
| | - Alessandro Ori
- Leibniz Institute on Aging Fritz Lipmann Institute (FLI), Beutenbergstraße 11, 07745 Jena, Germany; (S.M.M.R.); (E.C.); (A.O.); (F.N.)
| | - Francesco Neri
- Leibniz Institute on Aging Fritz Lipmann Institute (FLI), Beutenbergstraße 11, 07745 Jena, Germany; (S.M.M.R.); (E.C.); (A.O.); (F.N.)
| | - Rainer Quaas
- Chemicell GmbH, Erseburgstrasse 22–23, 12103 Berlin, Germany;
| | - Ingrid Hilger
- Institute of Diagnostic and Interventional Radiology, Jena University Hospital, Friedrich Schiller University Jena, Am Klinikum 1, D-07740 Jena, Germany;
- Correspondence: ; Tel.: +0049-3641-9325921
| |
Collapse
|
12
|
Archer M, Dogra N, Dovey Z, Ganta T, Jang HS, Khusid JA, Lantz A, Mihalopoulos M, Stockert JA, Zahalka A, Björnebo L, Gaglani S, Noh MR, Kaplan SA, Mehrazin R, Badani KK, Wiklund P, Tsao K, Lundon DJ, Mohamed N, Lucien F, Padanilam B, Gupta M, Tewari AK, Kyprianou N. Role of α- and β-adrenergic signaling in phenotypic targeting: significance in benign and malignant urologic disease. Cell Commun Signal 2021; 19:78. [PMID: 34284799 PMCID: PMC8290582 DOI: 10.1186/s12964-021-00755-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 05/28/2021] [Indexed: 01/17/2023] Open
Abstract
The urinary tract is highly innervated by autonomic nerves which are essential in urinary tract development, the production of growth factors, and the control of homeostasis. These neural signals may become dysregulated in several genitourinary (GU) disease states, both benign and malignant. Accordingly, the autonomic nervous system is a therapeutic target for several genitourinary pathologies including cancer, voiding dysfunction, and obstructing nephrolithiasis. Adrenergic receptors (adrenoceptors) are G-Protein coupled-receptors that are distributed throughout the body. The major function of α1-adrenoceptors is signaling smooth muscle contractions through GPCR and intracellular calcium influx. Pharmacologic intervention of α-and β-adrenoceptors is routinely and successfully implemented in the treatment of benign urologic illnesses, through the use of α-adrenoceptor antagonists. Furthermore, cell-based evidence recently established the antitumor effect of α1-adrenoceptor antagonists in prostate, bladder and renal tumors by reducing neovascularity and impairing growth within the tumor microenvironment via regulation of the phenotypic epithelial-mesenchymal transition (EMT). There has been a significant focus on repurposing the routinely used, Food and Drug Administration-approved α1-adrenoceptor antagonists to inhibit GU tumor growth and angiogenesis in patients with advanced prostate, bladder, and renal cancer. In this review we discuss the current evidence on (a) the signaling events of the autonomic nervous system mediated by its cognate α- and β-adrenoceptors in regulating the phenotypic landscape (EMT) of genitourinary organs; and (b) the therapeutic significance of targeting this signaling pathway in benign and malignant urologic disease. Video abstract.
Collapse
Affiliation(s)
- M. Archer
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
| | - N. Dogra
- Department of Pathology and Molecular and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Z. Dovey
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
| | - T. Ganta
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Division of Hematology and Medical Oncology, Mount Sinai Hospital, New York, NY USA
| | - H.-S. Jang
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
| | - J. A. Khusid
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
| | - A. Lantz
- Department of Molecular Medicine and Surgery, Section of Urology, Karolinska Institute, Stockholm, Sweden
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
| | - M. Mihalopoulos
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - J. A. Stockert
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
| | - A. Zahalka
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
| | - L. Björnebo
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
| | - S. Gaglani
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
| | - M. R. Noh
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
| | - S. A. Kaplan
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
| | - R. Mehrazin
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - K. K. Badani
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - P. Wiklund
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - K. Tsao
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Division of Hematology and Medical Oncology, Mount Sinai Hospital, New York, NY USA
| | - D. J. Lundon
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - N. Mohamed
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - F. Lucien
- Department of Urology, Mayo Clinic, Rochester, MN USA
| | - B. Padanilam
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
| | - M. Gupta
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
| | - A. K. Tewari
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - N. Kyprianou
- Department of Urology, Icahn School of Medicine at Mount Sinai, 6th Floor, 1425 Madison Avenue, New York, NY 10029 USA
- Department of Pathology and Molecular and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY USA
| |
Collapse
|
13
|
Sobol NT, Solernó LM, Beltrán B, Vásquez L, Ripoll GV, Garona J, Alonso DF. Anticancer activity of repurposed hemostatic agent desmopressin on AVPR2-expressing human osteosarcoma. Exp Ther Med 2021; 21:566. [PMID: 33850538 PMCID: PMC8027742 DOI: 10.3892/etm.2021.9998] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 12/03/2020] [Indexed: 01/15/2023] Open
Abstract
Osteosarcoma is the most prevalent primary bone malignancy. Due to its high aggressiveness, novel treatment strategies are urgently required to improve survival of patients with osteosarcoma, especially those with advanced disease. Desmopressin (dDAVP) is a widely used blood-saving agent that has been repurposed as an adjuvant agent for cancer management due to its antiangiogenic and antimetastatic properties. dDAVP acts as a selective agonist of the vasopressin membrane receptor type 2 (AVPR2) present in the microvascular endothelium and in some cancer cells, including breast, lung, colorectal and neuroendocrine tumor cells. Despite the fact that dDAVP has demonstrated its antitumor efficacy in a wide variety of tumor types, exploration of its potential anti-osteosarcoma activity has, to the best of our knowledge, not yet been conducted. Therefore, the aim of the present study was to evaluate the preclinical antitumor activity of dDAVP in osteosarcoma. Human MG-63 and U-2 OS osteosarcoma cell lines were used to assess in vitro and in vivo therapeutic effects of dDAVP. At low micromolar concentrations, dDAVP reduced AVPR2-expressing MG-63 cell growth in a concentration-dependent manner. In contrast, dDAVP exhibited no direct cytostatic effect on AVPR2-negative U-2 OS cells. As it would be expected for canonical AVPR2-activation, dDAVP raised intracellular cAMP levels in osteosarcoma cells, and coincubation with phosphodiesterase-inhibitor rolipram indicated synergistic antiproliferative activity. Cytostatic effects were associated with increased apoptosis, reduced mitotic index and impairment of osteosarcoma cell chemotaxis, as evaluated by TUNEL-labeling, mitotic body count in DAPI-stained cultures and Transwell migration assays. Intravenous administration of dDAVP (12 µg/kg; three times per week) to athymic mice bearing rapidly growing MG-63 xenografts, was indicated to be capable of reducing tumor progression after a 4-week treatment. No major alterations in animal weight, biochemical or hematological parameters were associated with dDAVP treatment, confirming its good tolerability and safety. Finally, AVPR2 expression was detected by immunohistochemistry in 66% of all evaluated chemotherapy-naive human conventional osteosarcoma biopsies. Taking these findings into account, repurposed agent dDAVP may represent an interesting therapeutic tool for the management of osteosarcoma. Further preclinical exploration of dDAVP activity on orthotopic or metastatic osteosarcoma models are required.
Collapse
Affiliation(s)
- Natasha Tatiana Sobol
- Center of Molecular and Translational Oncology, National University of Quilmes, Bernal B1876BXD, Buenos Aires, Argentina
| | - Luisina María Solernó
- Center of Molecular and Translational Oncology, National University of Quilmes, Bernal B1876BXD, Buenos Aires, Argentina
| | - Brady Beltrán
- Precision Medicine Research Center, School of Medicine, University of San Martín de Porres, Lima 15024, Perú
| | - Liliana Vásquez
- Precision Medicine Research Center, School of Medicine, University of San Martín de Porres, Lima 15024, Perú
| | - Giselle Vanina Ripoll
- Center of Molecular and Translational Oncology, National University of Quilmes, Bernal B1876BXD, Buenos Aires, Argentina
- National Council of Scientific and Technical Research (CONICET), Buenos Aires C1425FQB, Argentina
| | - Juan Garona
- Center of Molecular and Translational Oncology, National University of Quilmes, Bernal B1876BXD, Buenos Aires, Argentina
- National Council of Scientific and Technical Research (CONICET), Buenos Aires C1425FQB, Argentina
| | - Daniel Fernando Alonso
- Center of Molecular and Translational Oncology, National University of Quilmes, Bernal B1876BXD, Buenos Aires, Argentina
- National Council of Scientific and Technical Research (CONICET), Buenos Aires C1425FQB, Argentina
| |
Collapse
|
14
|
Iftikhar A, Islam M, Shepherd S, Jones S, Ellis I. Cancer and Stress: Does It Make a Difference to the Patient When These Two Challenges Collide? Cancers (Basel) 2021; 13:cancers13020163. [PMID: 33418900 PMCID: PMC7825104 DOI: 10.3390/cancers13020163] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/23/2020] [Accepted: 12/28/2020] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Head and neck cancers are the sixth most common cancer in the world. The burden of the disease has remained challenging over recent years despite the advances in treatments of other malignancies. The very use of the word malignancy brings about a stress response in almost all adult patients. Being told you have a tumour is not a word anyone wants to hear. We have embarked on a study which will investigate the effect of stress pathways on head and neck cancer patients and which signalling pathways may be involved. In the future, this will allow clinicians to better manage patients with head and neck cancer and reduce the patients’ stress so that this does not add to their tumour burden. Abstract A single head and neck Cancer (HNC) is a globally growing challenge associated with significant morbidity and mortality. The diagnosis itself can affect the patients profoundly let alone the complex and disfiguring treatment. The highly important functions of structures of the head and neck such as mastication, speech, aesthetics, identity and social interactions make a cancer diagnosis in this region even more psychologically traumatic. The emotional distress engendered as a result of functional and social disruption is certain to negatively affect health-related quality of life (HRQoL). The key biological responses to stressful events are moderated through the combined action of two systems, the hypothalamus–pituitary–adrenal axis (HPA) which releases glucocorticoids and the sympathetic nervous system (SNS) which releases catecholamines. In acute stress, these hormones help the body to regain homeostasis; however, in chronic stress their increased levels and activation of their receptors may aid in the progression of cancer. Despite ample evidence on the existence of stress in patients diagnosed with HNC, studies looking at the effect of stress on the progression of disease are scarce, compared to other cancers. This review summarises the challenges associated with HNC that make it stressful and describes how stress signalling aids in the progression of cancer. Growing evidence on the relationship between stress and HNC makes it paramount to focus future research towards a better understanding of stress and its effect on head and neck cancer.
Collapse
|
15
|
Sakakitani S, Podyma-Inoue KA, Takayama R, Takahashi K, Ishigami-Yuasa M, Kagechika H, Harada H, Watabe T. Activation of β2-adrenergic receptor signals suppresses mesenchymal phenotypes of oral squamous cell carcinoma cells. Cancer Sci 2020; 112:155-167. [PMID: 33007125 PMCID: PMC7780019 DOI: 10.1111/cas.14670] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 09/14/2020] [Accepted: 09/21/2020] [Indexed: 12/17/2022] Open
Abstract
Metastasis is a primary reason related to the mortality of oral squamous cell carcinoma (OSCC) patients. A program called epithelial-mesenchymal transition (EMT) has been shown to play a critical role in promoting metastasis in epithelium-derived carcinoma. During EMT, epithelial cancer cells acquire motile mesenchymal phenotypes and detach from primary tumors. Recent lines of evidence have suggested that EMT confers cancer cells with tumor-initiating ability. Therefore, selective targeting of EMT would lead to the development of effective therapeutic agents. In this study, using a chemical biology approach, we identified isoxsuprine, a β2-adrenergic receptor (β2-AR) agonist as a low-molecular-weight compound that interferes with the acquisition of mesenchymal phenotypes of oral cancer cells. Treatment of multiple types of oral cancer cells with isoxsuprine led to the downregulation of mesenchymal cell markers that was accompanied by reduced cell motility. Similar inhibitory effects were also observed for isoprenaline, a non-selective β-adrenergic receptor (β-AR) agonist. In addition, inhibition of cell migration upon treatment with isoxsuprine was reverted by a non-selective β-AR antagonist, propranolol, and the CRISPR/Cas9 system-mediated deletion of the β2-AR gene, suggesting that the effects exerted by isoxsuprine involved signals mediated by β2-AR. In addition, in a subcutaneous xenograft model of oral cancer cells, the administration of isoxsuprine effectively suppressed primary tumor growth, suggesting β2-AR signals to be a promising cancer therapeutic target for treatment of OSCC.
Collapse
Affiliation(s)
- Shintaro Sakakitani
- Department of Oral and Maxillofacial Surgery, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan.,Department of Biochemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Katarzyna A Podyma-Inoue
- Department of Biochemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Rina Takayama
- Department of Biochemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Kazuki Takahashi
- Department of Biochemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Mari Ishigami-Yuasa
- Department of Organic and Medicinal Chemistry, Chemical Biology Screening Center, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Hiroyuki Kagechika
- Department of Organic and Medicinal Chemistry, Chemical Biology Screening Center, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Hiroyuki Harada
- Department of Oral and Maxillofacial Surgery, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Tetsuro Watabe
- Department of Biochemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| |
Collapse
|
16
|
Agonist Effects of Propranolol on Non-Tumor Human Breast Cells. Cells 2020; 9:cells9041036. [PMID: 32331276 PMCID: PMC7226086 DOI: 10.3390/cells9041036] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 04/17/2020] [Accepted: 04/20/2020] [Indexed: 12/28/2022] Open
Abstract
The β-blocker propranolol (PROP) has been proposed as a repurposed treatment for breast cancer. The similarity of action between β-agonists and antagonists found on breast cells encouraged us to compare PROP and isoproterenol (ISO, agonist) signaling pathways on a human breast cell line. Cell proliferation was measured by cell counting and DNA-synthesis. Cell adhesion was measured counting the cells that remained adhered to the plastic after different treatments. Changes in actin cytoskeleton were observed by fluorescence staining and Western Blot. ISO and PROP caused a diminution of cell proliferation and an increase of cell adhesion, reverted by the pure β-antagonist ICI-118551. ISO and PROP induced a reorganization of actin cytoskeleton increasing F-actin, p-COFILIN and p-LIMK. While ISO elicited a marked enhancement of cAMP concentrations and an increase of vasodilator-stimulated phosphoprotein (VASP) and cAMP response element-binding protein (CREB) phosphorylation, PROP did not. Clathrin-mediated endocytosis inhibition or β-arrestin1 dominant-negative mutant abrogated PROP-induced cell adhesion and COFILIN phosphorylation. The fact that PROP has been proposed as an adjuvant drug for breast cancer makes it necessary to determine the specific action of PROP in breast models. These results provide an explanation for the discrepancies observed between experimental results and clinical evidence.
Collapse
|
17
|
Ghali GZ, Ghali MGZ. β adrenergic receptor modulated signaling in glioma models: promoting β adrenergic receptor-β arrestin scaffold-mediated activation of extracellular-regulated kinase 1/2 may prove to be a panacea in the treatment of intracranial and spinal malignancy and extra-neuraxial carcinoma. Mol Biol Rep 2020; 47:4631-4650. [PMID: 32303958 PMCID: PMC7165076 DOI: 10.1007/s11033-020-05427-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 04/03/2020] [Indexed: 12/03/2022]
Abstract
Neoplastically transformed astrocytes express functionally active cell surface β adrenergic receptors (βARs). Treatment of glioma models in vitro and in vivo with β adrenergic agonists variably amplifies or attenuates cellular proliferation. In the majority of in vivo models, β adrenergic agonists generally reduce cellular proliferation. However, treatment with β adrenergic agonists consistently reduces tumor cell invasive potential, angiogenesis, and metastasis. β adrenergic agonists induced decreases of invasive potential are chiefly mediated through reductions in the expression of matrix metalloproteinases types 2 and 9. Treatment with β adrenergic agonists also clearly reduce tumoral neoangiogenesis, which may represent a putatively useful mechanism to adjuvantly amplify the effects of bevacizumab. Bevacizumab is a monoclonal antibody targeting the vascular endothelial growth factor receptor. We may accordingly designate βagonists to represent an enhancer of bevacizumab. The antiangiogenic effects of β adrenergic agonists may thus effectively render an otherwise borderline effective therapy to generate significant enhancement in clinical outcomes. β adrenergic agonists upregulate expression of the major histocompatibility class II DR alpha gene, effectively potentiating the immunogenicity of tumor cells to tumor surveillance mechanisms. Authors have also demonstrated crossmodal modulation of signaling events downstream from the β adrenergic cell surface receptor and microtubular polymerization and depolymerization. Complex effects and desensitization mechanisms of the β adrenergic signaling may putatively represent promising therapeutic targets. Constant stimulation of the β adrenergic receptor induces its phosphorylation by β adrenergic receptor kinase (βARK), rendering it a suitable substrate for alternate binding by β arrestins 1 or 2. The binding of a β arrestin to βARK phosphorylated βAR promotes receptor mediated internalization and downregulation of cell surface receptor and contemporaneously generates a cell surface scaffold at the βAR. The scaffold mediated activation of extracellular regulated kinase 1/2, compared with protein kinase A mediated activation, preferentially favors cytosolic retention of ERK1/2 and blunting of nuclear translocation and ensuant pro-transcriptional activity. Thus, βAR desensitization and consequent scaffold assembly effectively retains the cytosolic homeostatic functions of ERK1/2 while inhibiting its pro-proliferative effects. We suggest these mechanisms specifically will prove quite promising in developing primary and adjuvant therapies mitigating glioma growth, angiogenesis, invasive potential, and angiogenesis. We suggest generating compounds and targeted mutations of the β adrenergic receptor favoring β arrestin binding and scaffold facilitated activation of ERK1/2 may hold potential promise and therapeutic benefit in adjuvantly treating most or all cancers. We hope our discussion will generate fruitful research endeavors seeking to exploit these mechanisms.
Collapse
Affiliation(s)
- George Zaki Ghali
- United States Environmental Protection Agency, Arlington, VA, USA.,Emeritus Professor, Department of Toxicology, Purdue University, West Lafayette, IN, USA
| | - Michael George Zaki Ghali
- Department of Neurological Surgery, University of California, San Francisco, 505 Parnassus Avenue, Box-0112, San Francisco, CA, 94143, USA. .,Department of Neurological Surgery, Karolinska Institutet, Nobels väg 6, Solna and Alfred Nobels Allé 8, Huddinge, SE-171 77, Stockholm, Sweden.
| |
Collapse
|
18
|
β2-Adrenergic Signalling Promotes Cell Migration by Upregulating Expression of the Metastasis-Associated Molecule LYPD3. BIOLOGY 2020; 9:biology9020039. [PMID: 32098331 PMCID: PMC7168268 DOI: 10.3390/biology9020039] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 02/13/2020] [Accepted: 02/20/2020] [Indexed: 12/24/2022]
Abstract
Metastasis is associated with poor prognosis in breast cancer. Although some studies suggest beta-blockers increase survival by delaying metastasis, others have been discordant. This study provides both insights into the anomalous findings and identifies potential biomarkers that may be treatment targets. Cell line models of basal-type and oestrogen receptor-positive breast cancer were profiled for basal levels of adrenoceptor gene/protein expression, and β2-adrenoceptor mediated cell behaviour including migration, invasion, adhesion, and survival in response to adrenoceptor agonist/antagonist treatment. Protein profiling and histology identified biomarkers and drug targets. Baseline levels of adrenoceptor gene expression are higher in basal-type rather than oestrogen receptor-positive cancer cells. Norepinephrine (NE) treatment increased invasive capacity in all cell lines but did not increase proliferation/survival. Protein profiling revealed the upregulation of the pro-metastatic gene Ly6/PLAUR Domain-Containing Protein 3 (LYPD3) in norepinephrine-treated MDA-MB-468 cells. Histology confirmed selective LYPD3 expression in primary and metastatic breast tumour samples. These findings demonstrate that basal-type cancer cells show a more aggressive adrenoceptor-β2-activated phenotype in the resting and stimulated state, which is attenuated by adrenoceptor-β2 inhibition. This study also highlights the first association between ADRβ2 signalling and LYPD3; its knockdown significantly reduced the basal and norepinephrine-induced activity of MCF-7 cells in vitro. The regulation of ADRβ2 signalling by LYPD3 and its metastasis promoting activities, reveal LYPD3 as a promising therapeutic target in the treatment of breast and other cancers.
Collapse
|
19
|
Garona J, Pifano M, Ripoll G, Alonso DF. Development and therapeutic potential of vasopressin synthetic analog [V 4Q 5]dDAVP as a novel anticancer agent. VITAMINS AND HORMONES 2020; 113:259-289. [PMID: 32138951 DOI: 10.1016/bs.vh.2019.08.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Since its discovery, arginine vasopressin (AVP) was subjected to several modifications with the aim of obtaining novel derivatives with increased potency and selectivity for biomedical use. Desmopressin (dDAVP) is a first generation synthetic analog of AVP with hemostatic and antimetastatic activity. dDAVP acts as a selective agonist of the arginine vasopressin type 2 receptor (AVPR2) present in microvascular endothelium and cancer cells. Considering its selective effects on AVPR2-expressing malignant and vascular tissue, and interesting antitumor profile, dDAVP was used as a lead compound for the development of novel peptide analogs with enhanced anticancer efficacy. After conducting different structure-activity relationship studies to determine key aminoacidic positions for its antitumor activity against AVPR2-expressing malignant cells, dDAVP was rationally modified and a wide panel of synthetic analogs with different sequence and structural modifications was assessed. As a result of this structure-based drug derivatization novel AVP analog [V4Q5]dDAVP (1-deamino-4-valine-5-glutamine-8-d-arginine vasopressin) was selected as the most active candidate and further developed. [V4Q5]dDAVP was evaluated in highly aggressive and metastatic cancer preclinical models deploying enhanced cytostatic, antimetastatic and angiostatic effects in comparison to parental peptide dDAVP. In addition, novel compound demonstrated good tolerability as evaluated in several toxicological studies, and cooperative therapeutic effects after combination with standard-of-care chemotherapy. In summary, due to its ability to inhibit growth and tumor-associated angiogenesis, as well as impairing progression of metastatic disease, AVP analogs such as novel [V4Q5]dDAVP are promising compounds for further development as coadjuvant agents for the management of advance or recurrent cancers.
Collapse
Affiliation(s)
- Juan Garona
- Laboratory of Molecular Oncology, Science and Technology Department, National University of Quilmes, Buenos Aires, Argentina.
| | - Marina Pifano
- Laboratory of Molecular Oncology, Science and Technology Department, National University of Quilmes, Buenos Aires, Argentina
| | - Giselle Ripoll
- Laboratory of Molecular Oncology, Science and Technology Department, National University of Quilmes, Buenos Aires, Argentina
| | - Daniel F Alonso
- Laboratory of Molecular Oncology, Science and Technology Department, National University of Quilmes, Buenos Aires, Argentina
| |
Collapse
|
20
|
Ha M, Kim DW, Kim J, Hong CM, Park SM, Woo IA, Kim MY, Koo H, Namkoong J, Kim J, Han ME, Song P, Hur J, Kang CD, Kim YH, Lee D, Oh SO. Prognostic role of the beta-2 adrenergic receptor in clear cell renal cell carcinoma. Anim Cells Syst (Seoul) 2019; 23:365-369. [PMID: 31700702 PMCID: PMC6830282 DOI: 10.1080/19768354.2019.1658638] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 07/31/2019] [Accepted: 08/14/2019] [Indexed: 12/18/2022] Open
Abstract
The beta-2 adrenergic receptor (ADRB2) regulates the proliferation, apoptosis, angiogenesis, migration, and metastasis of cancer cells. However, its function in the progression of clear cell renal cell carcinoma (ccRCC) is unknown. Here, we report that ADRB2 can be a novel prognostic factor for patients with ccRCC. The differential expression of ADRB2 in low-stage (stages I and II), high-stage (stages III and IV), low-grade (grades I and II), and high-grade (grades III and IV) ccRCC was identified in cohorts of patients from The Cancer Genome Atlas and the International Cancer Genome Consortium. We evaluated ADRB2 expression as a prognostic factor using the Kaplan-Meier survival curve, multivariate analysis, time-dependent area under the curve (AUC) of Uno’s C-index, and AUC of the receiver operating characteristics (ROC) at five years. Kaplan-Meier analysis revealed that reduced ADRB2 expression is associated with poor prognosis in ccRCC patients. Analysis of C-indices and AUC-ROC further confirmed this result. Moreover, multivariate analysis confirmed the prognostic significance of ADRB2 expression. Collectively, these findings suggest that ADRB2 is a potential prognostic factor for ccRCC.
Collapse
Affiliation(s)
- Mihyang Ha
- Department of Anatomy, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Dong Woo Kim
- Department of Premedicine, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Jayoung Kim
- Department of Convergence Medicine, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Chae Mi Hong
- Department of Convergence Medicine, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Su Min Park
- Department of Convergence Medicine, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - In Ae Woo
- Department of Convergence Medicine, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Min Yong Kim
- Department of Convergence Medicine, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Hyunjun Koo
- Department of Premedicine, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Jin Namkoong
- Department of Premedicine, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Jaehyun Kim
- Department of Premedicine, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Myoung-Eun Han
- Department of Anatomy, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Parkyong Song
- Department of Convergence Medicine, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Jin Hur
- Department of Convergence Medicine, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Chi-Dug Kang
- Department of Convergence Medicine, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Yun Hak Kim
- Department of Anatomy, Department of Biomedical Informatics, and Biomedical Research Institute, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Dongjun Lee
- Department of Convergence Medicine, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Sae-Ock Oh
- Department of Anatomy, Pusan National University School of Medicine, Yangsan, Republic of Korea
| |
Collapse
|
21
|
Rivero EM, Martinez LM, Bruque CD, Gargiulo L, Bruzzone A, Lüthy IA. Prognostic significance of α- and β2-adrenoceptor gene expression in breast cancer patients. Br J Clin Pharmacol 2019; 85:2143-2154. [PMID: 31218733 DOI: 10.1111/bcp.14030] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 04/04/2019] [Accepted: 05/21/2019] [Indexed: 12/12/2022] Open
Abstract
AIMS Breast cancer is the most frequently diagnosed and leading cause of cancer death among women worldwide. It was classified within molecular intrinsic subtypes: luminal A, luminal B, human epidermal growth factor receptor 2-enriched and basal-like. Epinephrine and norepinephrine, released during stress, bind to adrenoceptors. α2 -adrenoceptors are encoded by the ADRA2A, ADRA2B and ADRA2C genes and β2 by ADRB2. METHODS We compiled several publicly available Affymetrix gene expression datasets, obtaining a large cohort of 1924 patients with distant metastasis-free survival (DMFS) data and evaluated the association between adrenoceptor expression, clinicopathological markers and outcome. RESULTS ADRA2A high expressing tumours also expressed hormone receptors and presented diminished tumour size, grade and not compromised lymph nodes. ADRB2 high expression was found in smaller, low grade, oestrogen receptor-positive tumours. Both were significantly associated with the absence of metastasis. High expression of ADRA2C was positively associated with increased tumour size and metastatic relapse. We observed a significant increase in DMFS of patients with high ADRA2A (hazard ratio 0.54, 95% CI 0.45-0.65, P < .001) and ADRB2 (0.77, 0.64-0.93, P = .006) expression and a decrease with ADRA2C high expression (1.45, 1.16-1.81, P = .001). For patients with luminal tumours, ADRA2A was the only factor that retained its significance as an independent predictor of DMFS while ADRA2C expression was an independent predictor for worse prognosis in basal-like tumours. CONCLUSIONS We herein provide new insight for a potential role of ADRA2A and ADRA2C in breast cancer. In low- and medium-income countries, their incorporation to routine immunohistochemistry analysis of biopsies or tumour samples, could provide additional low-cost prognostic factors.
Collapse
Affiliation(s)
- Ezequiel Mariano Rivero
- Laboratory of Hormones and Cancer, Instituto de Biología y Medicina Experimental (IBYME) - CONICET, Ciudad Autónoma de Buenos Aires, Argentina
| | | | - Carlos David Bruque
- ANLIS, Centro Nacional de Genética Médica, Ciudad Autónoma de Buenos Aires, Argentina
| | - Lucia Gargiulo
- Laboratory of Hormones and Cancer, Instituto de Biología y Medicina Experimental (IBYME) - CONICET, Ciudad Autónoma de Buenos Aires, Argentina
| | - Ariana Bruzzone
- Instituto de Investigaciones Bioquímicas Bahía Blanca INIBIBB-CONICET, Bahía Blanca, Argentina
| | - Isabel Alicia Lüthy
- Laboratory of Hormones and Cancer, Instituto de Biología y Medicina Experimental (IBYME) - CONICET, Ciudad Autónoma de Buenos Aires, Argentina
| |
Collapse
|
22
|
Kim SH, Yi SJ, Lee H, Kim JH, Oh MJ, Song EJ, Kim K, Jhun BH. β 2-Adrenergic receptor (β 2-AR) agonist formoterol suppresses differentiation of L6 myogenic cells by blocking PI3K-AKT pathway. Anim Cells Syst (Seoul) 2019; 23:18-25. [PMID: 30834155 PMCID: PMC6394304 DOI: 10.1080/19768354.2018.1561516] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 11/20/2018] [Accepted: 12/05/2018] [Indexed: 01/01/2023] Open
Abstract
β2-Adrenergic receptor (β2-AR) is implicated in muscle metabolic activities such as glycogen metabolism, glucose uptake, lipolysis and muscle growth. However, the functional role of β2-AR in the differentiation of skeletal muscle is largely unknown. Here, we examined the functional role of β2-AR in L6 myoblast differentiation using the long-term-acting β2-AR-specific agonist formoterol. We observed that formoterol treatment strongly suppressed L6 myoblast differentiation and the expression of myosin heavy chain (MHC) in a dose- and time-dependent manner. Showing that both long-acting agonist (formoterol) and short-acting agonist (terbutaline) inhibited the induction of MHC protein, whereas β2-AR antagonist (ICI-118,551) upregulated MHC expression, we clearly demonstrated that β2-AR is involved in L6 myoblast differentiation. Furthermore, our pharmacological inhibition study revealed that the PI3K–AKT pathway is the main signaling pathway for myotube formation. Formoterol inhibited the activation of PI3K–AKT signaling, but not that of ERK signaling. Moreover, formoterol selectively inhibited AKT activation by IGF-I, but not by insulin. Collectively, our findings reveal a previously undocumented role of β2-AR activation in modulating the differentiation of L6 myoblasts.
Collapse
Affiliation(s)
- So-Hyeon Kim
- Department of Cogno-Mechatronics Engineering, Pusan National University, Busan, Republic of Korea
| | - Sun-Ju Yi
- School of Biological Sciences, College of Natural Sciences, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Hyerim Lee
- School of Biological Sciences, College of Natural Sciences, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Ji-Hyun Kim
- Department of Cogno-Mechatronics Engineering, Pusan National University, Busan, Republic of Korea
| | - Myung-Ju Oh
- Department of Cogno-Mechatronics Engineering, Pusan National University, Busan, Republic of Korea
| | - Eun-Ju Song
- Department of Cogno-Mechatronics Engineering, Pusan National University, Busan, Republic of Korea
| | - Kyunghwan Kim
- School of Biological Sciences, College of Natural Sciences, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Byung H Jhun
- Department of Cogno-Mechatronics Engineering, Pusan National University, Busan, Republic of Korea
| |
Collapse
|
23
|
Solomon I, Voiculescu VM, Caruntu C, Lupu M, Popa A, Ilie MA, Albulescu R, Caruntu A, Tanase C, Constantin C, Neagu M, Boda D. Neuroendocrine Factors and Head and Neck Squamous Cell Carcinoma: An Affair to Remember. DISEASE MARKERS 2018; 2018:9787831. [PMID: 29854027 PMCID: PMC5966665 DOI: 10.1155/2018/9787831] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 03/21/2018] [Accepted: 04/15/2018] [Indexed: 02/07/2023]
Abstract
Head and neck squamous cell carcinoma (HNSCC) is one of the most aggressive malignancies. Therefore, the major goal of cancer treatment is inhibition of tumor cell growth and of metastasis development. In order to choose the best management option for HNSCC patients, we need to identify reliable prognostic factors and to develop new molecular techniques in order to obtain a better understanding of therapy resistance. By acting as neurohormones, neurotransmitters, or neuromodulators, the neuroendocrine factors are able to signal the maintenance of physiological homeostasis or progression to malignant disease. Certain neuropeptides possess strong antitumor properties acting as tumor suppressors and immunomodulators, providing additional benefits for future potential therapeutic strategies. In light of the current understanding, cancer starts as a localized disease that can be effectively treated if discovered on proper time. Unfortunately, more than often cancer cells migrate to the surrounding tissues generating distant metastases, thus making the prognosis and survival in this stage much worse. As cellular migration is mandatory for tumor invasion and metastasis development, searching for alternate controllers of these processes, such as the neuroendocrine factors, it is an active tremendous task.
Collapse
Affiliation(s)
- Iulia Solomon
- 1Department of Dermatology and Allergology, Elias Emergency University Hospital, Bucharest, Romania
| | - Vlad Mihai Voiculescu
- 1Department of Dermatology and Allergology, Elias Emergency University Hospital, Bucharest, Romania
- 2Department of Dermatology, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Constantin Caruntu
- 3Department of Physiology, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
- 4Department of Dermatology, “Prof. N. C. Paulescu” National Institute of Diabetes, Nutrition and Metabolic Diseases, Bucharest, Romania
| | - Mihai Lupu
- 5Department of Dermatology, MEDAS Titan Medical Center, Bucharest, Romania
| | - Alexandra Popa
- 1Department of Dermatology and Allergology, Elias Emergency University Hospital, Bucharest, Romania
| | - Mihaela Adriana Ilie
- 6Dermatology Research Laboratory, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
- 7Department of Biochemistry, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Radu Albulescu
- 8Chemical and Pharmaceutical National Institute, Bucharest, Romania
| | - Ana Caruntu
- 9Department of Oral and Maxillofacial Surgery, Carol Davila Central Military Emergency Hospital, Bucharest, Romania
- 10Faculty of Medicine, Titu Maiorescu University, Bucharest, Romania
| | - Cristiana Tanase
- 10Faculty of Medicine, Titu Maiorescu University, Bucharest, Romania
- 11Victor Babes National Institute of Pathology, Bucharest, Romania
| | - Carolina Constantin
- 11Victor Babes National Institute of Pathology, Bucharest, Romania
- 12Colentina Clinical Hospital, Bucharest, Romania
| | - Monica Neagu
- 11Victor Babes National Institute of Pathology, Bucharest, Romania
- 12Colentina Clinical Hospital, Bucharest, Romania
- 13Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Daniel Boda
- 6Dermatology Research Laboratory, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| |
Collapse
|
24
|
Delayed neurochemical effects of prenatal exposure to MeHg in the cerebellum of developing rats. Toxicol Lett 2017; 284:161-169. [PMID: 29258870 DOI: 10.1016/j.toxlet.2017.12.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 12/03/2017] [Accepted: 12/09/2017] [Indexed: 01/09/2023]
Abstract
Human fetuses and neonates are particularly vulnerable to methylmercury (MeHg)-induced brain damage and are sensitive even to low exposure levels. Previous work of our group evidence that prenatal exposure to MeHg causes cognitive and behavioral alterations and disrupt hippocampus signaling. The current study aimed to investigate the effect of gestational exposure of rats to MeHg at low doses (1 or 2 mg/kg) on parameters of redox imbalance and key signaling pathways in the cerebellum of their offspring. Pregnant females received MeHg (treated group) or 0.9% saline water (control group) by gavage in alternated days from gestational day 5 (GD5) until parturition and analyzes were proceed in the cerebellum of 30-day-old pups. We found increased lipid peroxidation and protein carbonylation levels as well as decreased SH content in pups prenatally exposed to 2 mg/kg MeHg. In addition, misregulated SOD/catalase activities supported imbalanced redox equilibrium. We found decreased GSK3β(Ser9) phosphorylation, suggesting activation of this enzyme and dephosphorylation/inhibition of ERK1/2 and JNK pathways. Increased PKAα catalytic subunit could be upstream of hyperphosphorylated c-Raf(Ser259) and downregulated MAPK pathway. In addition, we found raised levels of the Ca2+-dependent protein phosphatase 2 B (PP2B). We also found preserved immunohistochemical staining for both glial fibrillary acidic protein (GFAP) and NeuN in MeHg-exposed pups. Western blot analysis showed unaltered levels of BAX/BCL-XL, BAD/BCL-2 and active caspase 3. Together, these findings support absence of reactive astrocytes, neuronal damage and apoptotic cell death in the cerebellum of MeHg treated pups. The present study provides evidence that prenatal exposure to MeHg leads to later redox imbalance and disrupted signaling mechanisms in the cerebellum of 30-day-old pups potentially predisposing them to long-lasting neurological impairments in CNS.
Collapse
|
25
|
Tuglu MM, Bostanabad SY, Ozyon G, Dalkiliç B, Gurdal H. The role of dual‑specificity phosphatase 1 and protein phosphatase 1 in β2‑adrenergic receptor‑mediated inhibition of extracellular signal regulated kinase 1/2 in triple negative breast cancer cell lines. Mol Med Rep 2017; 17:2033-2043. [PMID: 29257221 DOI: 10.3892/mmr.2017.8092] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 10/24/2017] [Indexed: 11/06/2022] Open
Abstract
Triple negative breast cancer cell lines express high levels of β2-adrenergic receptor, which have a significant influence on the activity of extracellular signal‑regulated kinase (ERK)1/2. Therefore, it is important to understand the link between β2‑adrenergic receptor signaling and ERK1/2 activity in terms of cancer cell regulation and cancer progression. Although the molecular mechanisms are not completely clarified, β2‑adrenergic receptor stimulation appears to reduce the basal levels of phosphorylated (p)ERK1/2 in MDA‑MB‑231 breast cancer cells. The aim of the current study was to determine the mechanism of β2‑adrenergic receptor‑mediated ERK1/2 dephosphorylation by investigating the role of dual‑specificity phosphatase (DUSP)1/6 and protein phosphatase (PP)1/2, which are established regulators of ERK1/2 phosphorylation, in MDA‑MB‑231 and MDA‑MB‑468 breast cancer cell lines. (E)‑2‑benzylidene‑3‑(cyclohexylamino)‑2,3‑ dihydro‑1H‑inden‑1‑one (BCI) and calyculin A were employed as DUSP1/6 and PP1/PP2 inhibitors, respectively. Subsequently, the protein levels of DUSP1, PP1, pPP1, ERK1/2 and pERK1/2 were measured by western blot analysis. Cells were transfected with DUSP1 small interfering (si)RNA or PP1 siRNA to inhibit their expression. The results demonstrated that β2‑adrenergic receptor agonists led to the dephosphorylation of basal pERK1/2 in MDA‑MB‑231 and MDA‑MB‑468 cells. The DUSP1/6 inhibitor, BCI, and the PP1/PP2 inhibitor, calyculin A, antagonized the β2‑adrenergic receptor‑mediated dephosphorylation of ERK1/2. Furthermore, β2‑adrenergic receptor stimulation increased the protein expression level of DUSP1, with no effects on DUSP6, PP1 and PP2 expression, and enhanced the expression of the active form of PP1. Downregulation of the expression of DUSP1 or PP1 led to a decline in the β2‑adrenergic receptor‑mediated dephosphorylation of ERK1/2. The results of the present study indicate that β2‑adrenergic receptor‑mediated dephosphorylation of ERK1/2 may be associated with the activity of DUSP1 and PP1 in MDA‑MB‑231 and MDA‑MB‑468 triple negative breast cancer cell lines. The clinical importance of β2‑adrenergic receptor‑mediated inactivation of ERK1/2 as well as the activation of DUSP1 and PP1 should be carefully evaluated in future studies, particularly when β2‑adrenergic blockers are used in patients with triple negative breast cancer.
Collapse
Affiliation(s)
- Matilda Merve Tuglu
- Department of Medical Pharmacology, Faculty of Medicine, University of Ankara, 06100 Ankara, Turkey
| | | | - Gozde Ozyon
- Biotechnology Institute of Ankara University, 06110 Ankara, Turkey
| | - Başak Dalkiliç
- Department of Medical Pharmacology, Faculty of Medicine, University of Ankara, 06100 Ankara, Turkey
| | - Hakan Gurdal
- Department of Medical Pharmacology, Faculty of Medicine, University of Ankara, 06100 Ankara, Turkey
| |
Collapse
|
26
|
He JJ, Zhang WH, Liu SL, Chen YF, Liao CX, Shen QQ, Hu P. Activation of β-adrenergic receptor promotes cellular proliferation in human glioblastoma. Oncol Lett 2017; 14:3846-3852. [PMID: 28927156 DOI: 10.3892/ol.2017.6653] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 04/13/2017] [Indexed: 12/23/2022] Open
Abstract
Glioblastoma multiforme is the most common and aggressive form of primary malignant brain tumor. Previous evidence demonstrates that β-adrenergic receptors (β-ARs) are closely associated with the occurrence and development of brain tumors. However, the functional role of β-ARs in human glioblastoma and the underlying mechanisms are not fully understood. In the present study, by using the MTT assay, western blotting, and the reverse transcription polymerase chain reaction, it was revealed that isoproterenol (ISO), an agonist of β-ARs, promoted the proliferation of U251 cells but not U87-MG cells, and that this effect was blocked by the β-ARs antagonist propranolol. It was also demonstrated that ISO transiently induced extracellular signal-related kinase 1/2 (ERK1/2) phosphorylation, and that blocking the mitogen-activated protein kinase pathway by U0126 inhibited ERK1/2 phosphorylation and suppressed U251 cell proliferation. In addition, β-ARs activation increased the expression of matrix metalloproteinase (MMP) family members MMP-2 and MMP-9 mRNA through ERK1/2 activation. In conclusion, these data suggest that β-ARs induce ERK1/2 phosphorylation, which may in turn increase MMPs expression to promote U251 cell proliferation. These results provide additional insight into the specific roles of β-ARs in glioblastoma.
Collapse
Affiliation(s)
- Jing-Jing He
- Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi 330001, P.R. China
| | - Wen-Hua Zhang
- Institute of Life Science, Nanchang University, Nanchang, Jiangxi 330031, P.R. China
| | - Shi-Ling Liu
- Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi 330001, P.R. China
| | - Yi-Fang Chen
- Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi 330001, P.R. China
| | - Chen-Xi Liao
- Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi 330001, P.R. China
| | - Qian-Qing Shen
- Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi 330001, P.R. China
| | - Ping Hu
- Institute of Translational Medicine, Nanchang University, Nanchang, Jiangxi 330001, P.R. China
| |
Collapse
|
27
|
Wnorowski A. To block it, or not to block it? J Cancer Res Clin Oncol 2017; 143:2631-2633. [PMID: 28349195 PMCID: PMC5693973 DOI: 10.1007/s00432-017-2400-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 03/14/2017] [Indexed: 02/08/2023]
Affiliation(s)
- Artur Wnorowski
- Department of Biopharmacy, Medical University of Lublin, Collegium Pharmaceuticum, Chodzki 4a01a, 20-093, Lublin, Poland.
| |
Collapse
|
28
|
Gargiulo L, May M, Rivero EM, Copsel S, Lamb C, Lydon J, Davio C, Lanari C, Lüthy IA, Bruzzone A. A Novel Effect of β-Adrenergic Receptor on Mammary Branching Morphogenesis and its Possible Implications in Breast Cancer. J Mammary Gland Biol Neoplasia 2017; 22:43-57. [PMID: 28074314 DOI: 10.1007/s10911-017-9371-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 01/02/2017] [Indexed: 12/20/2022] Open
Abstract
Understanding the mechanisms that govern normal mammary gland development is crucial to the comprehension of breast cancer etiology. β-adrenergic receptors (β-AR) are targets of endogenous catecholamines such as epinephrine that have gained importance in the context of cancer biology. Differences in β2-AR expression levels may be responsible for the effects of epinephrine on tumor vs non-tumorigenic breast cell lines, the latter expressing higher levels of β2-AR. To study regulation of the breast cell phenotype by β2-AR, we over-expressed β2-AR in MCF-7 breast cancer cells and knocked-down the receptor in non-tumorigenic MCF-10A breast cells. In MCF-10A cells having knocked-down β2-AR, epinephrine increased cell proliferation and migration, similar to the response by tumor cells. In contrast, in MCF-7 cells overexpressing the β2-AR, epinephrine decreased cell proliferation and migration and increased adhesion, mimicking the response of the non-tumorigenic MCF-10A cells, thus underscoring that β2-AR expression level is a key player in cell behavior. β-adrenergic stimulation with isoproterenol induced differentiation of breast cells growing in 3-dimension cell culture, and also the branching of murine mammary epithelium in vivo. Branching induced by isoproterenol was abolished in fulvestrant or tamoxifen-treated mice, demonstrating that the effect of β-adrenergic stimulation on branching is dependent on the estrogen receptor (ER). An ER-independent effect of isoproterenol on lumen architecture was nonetheless found. Isoproterenol significantly increased the expression of ERα, Ephrine-B1 and fibroblast growth factors in the mammary glands of mice, and in MCF-10A cells. In a poorly differentiated murine ductal carcinoma, isoproterenol also decreased tumor growth and induced tumor differentiation. This study highlights that catecholamines, through β-AR activation, seem to be involved in mammary gland development, inducing mature duct formation. Additionally, this differentiating effect could be resourceful in a breast tumor context.
Collapse
Affiliation(s)
- Lucía Gargiulo
- Instituto de Biología y Medicina Experimental-CONICET, Vuelta de Obligado 2490, C1428ADN, Buenos Aires, CABA, Argentina
| | - María May
- Instituto de Biología y Medicina Experimental-CONICET, Vuelta de Obligado 2490, C1428ADN, Buenos Aires, CABA, Argentina
| | - Ezequiel M Rivero
- Instituto de Biología y Medicina Experimental-CONICET, Vuelta de Obligado 2490, C1428ADN, Buenos Aires, CABA, Argentina
| | - Sabrina Copsel
- Instituto de Biología y Medicina Experimental-CONICET, Vuelta de Obligado 2490, C1428ADN, Buenos Aires, CABA, Argentina
- Laboratorio de Farmacología de Receptores, Departamento de Farmacología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junin 956, 1113, Buenos Aires, CABA, Argentina
| | - Caroline Lamb
- Instituto de Biología y Medicina Experimental-CONICET, Vuelta de Obligado 2490, C1428ADN, Buenos Aires, CABA, Argentina
| | - John Lydon
- Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Carlos Davio
- Laboratorio de Farmacología de Receptores, Departamento de Farmacología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junin 956, 1113, Buenos Aires, CABA, Argentina
| | - Claudia Lanari
- Instituto de Biología y Medicina Experimental-CONICET, Vuelta de Obligado 2490, C1428ADN, Buenos Aires, CABA, Argentina
| | - Isabel A Lüthy
- Instituto de Biología y Medicina Experimental-CONICET, Vuelta de Obligado 2490, C1428ADN, Buenos Aires, CABA, Argentina
| | - Ariana Bruzzone
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), CONICET-Universidad Nacional del Sur, Camino La Carrindanga km 7, 8000, Bahía Blanca, Argentina.
| |
Collapse
|
29
|
Pifano M, Garona J, Capobianco CS, Gonzalez N, Alonso DF, Ripoll GV. Peptide Agonists of Vasopressin V2 Receptor Reduce Expression of Neuroendocrine Markers and Tumor Growth in Human Lung and Prostate Tumor Cells. Front Oncol 2017; 7:11. [PMID: 28194370 PMCID: PMC5276816 DOI: 10.3389/fonc.2017.00011] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 01/11/2017] [Indexed: 12/14/2022] Open
Abstract
Neuroendocrine tumors (NETs) comprise a heterogeneous group of malignancies that express neuropeptides as synaptophysin, chromogranin A (CgA), and specific neuronal enolase (NSE), among others. Vasopressin (AVP) is a neuropeptide with an endocrine, paracrine, and autocrine effect in normal and pathological tissues. AVP receptors are present in human lung, breast, pancreatic, colorectal, and gastrointestinal tumors. While AVP V1 receptors are associated with stimulation of cellular proliferation, AVP V2 receptor (V2r) is related to antiproliferative effects. Desmopressin (dDAVP) is a synthetic analog of AVP that acts as a selective agonist for the V2r, which shows antitumor properties in breast and colorectal cancer models. Recently, we developed a derivative of dDAVP named [V4Q5]dDAVP, which presents higher antitumor effects in a breast cancer model compared to the parental compound. The goal of present work was to explore the antitumor properties of the V2r agonist dDAVP and its novel analog [V4Q5]dDAVP on aggressive human lung (NCI-H82) and prostate cancer (PC-3) cell lines with neuroendocrine (NE) characteristics. We study the presence of specific NE markers (CgA and NSE) and V2r expression in NCI-H82 and PC-3. Both cell lines express high levels of NE markers NSE and CgA but then incubation with dDAVP diminished expression levels of both markers. DDAVP and [V4Q5]dDAVP significantly reduced proliferation, doubling time, and migration in both tumor cell cultures. [V4Q5]dDAVP analog showed a higher cytostatic effect than dDAVP, on cellular proliferation in the NCI-H82 cell line. Silencing of V2r using small interfering RNA significantly attenuated the inhibitory effects of [V4Q5]dDAVP on NCI-H82 cell proliferation. We, preliminarily, explored the in vivo effect of dDAVP and [V4Q5]dDAVP on NCI-H82 small cell lung cancer xenografts. Treated tumors (0.3 μg kg-1, thrice a week) grew slower in comparison to vehicle-treated animals. In this work, we demonstrated that the specific agonists of V2r, dDAVP, and [V4Q5]dDAVP displays antitumor capacity on different human models of lung and prostate cancers with NE features, showing their potential therapeutic benefits in the treatment of these aggressive tumors.
Collapse
Affiliation(s)
- Marina Pifano
- Laboratory of Molecular Oncology, Quilmes National University , Bernal, Buenos Aires , Argentina
| | - Juan Garona
- Laboratory of Molecular Oncology, Quilmes National University , Bernal, Buenos Aires , Argentina
| | - Carla S Capobianco
- Laboratory of Molecular Oncology, Quilmes National University , Bernal, Buenos Aires , Argentina
| | - Nazareno Gonzalez
- Laboratory of Molecular Oncology, Quilmes National University , Bernal, Buenos Aires , Argentina
| | - Daniel F Alonso
- Laboratory of Molecular Oncology, Quilmes National University , Bernal, Buenos Aires , Argentina
| | - Giselle V Ripoll
- Laboratory of Molecular Oncology, Quilmes National University , Bernal, Buenos Aires , Argentina
| |
Collapse
|
30
|
Pagano F, Angelini F, Siciliano C, Tasciotti J, Mangino G, De Falco E, Carnevale R, Sciarretta S, Frati G, Chimenti I. Beta2-adrenergic signaling affects the phenotype of human cardiac progenitor cells through EMT modulation. Pharmacol Res 2017; 127:41-48. [PMID: 28099883 DOI: 10.1016/j.phrs.2017.01.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 12/12/2016] [Accepted: 01/13/2017] [Indexed: 01/08/2023]
Abstract
Human cardiac progenitor cells (CPCs) offer great promises to cardiac cell therapy for heart failure. Many in vivo studies have shown their therapeutic benefits, paving the way for clinical translation. The 3D model of cardiospheres (CSs) represents a unique niche-like in vitro microenvironment, which includes CPCs and supporting cells. CSs have been shown to form through a process mediated by epithelial-to-mesenchymal transition (EMT). β2-Adrenergic signaling significantly affects stem/progenitor cells activation and mobilization in multiple tissues, and crosstalk between β2-adrenergic signaling and EMT processes has been reported. In the present study, we aimed at investigating the biological response of CSs to β2-adrenergic stimuli, focusing on EMT modulation in the 3D culture system of CSs. We treated human CSs and CS-derived cells (CDCs) with the β2-blocker butoxamine (BUT), using either untreated or β2 agonist (clenbuterol) treated CDCs as control. BUT-treated CS-forming cells displayed increased migration capacity and a significant increase in their CS-forming ability, consistently associated with increased expression of EMT-related genes, such as Snai1. Moreover, long-term BUT-treated CDCs contained a lower percentage of CD90+ cells, and this feature has been previously correlated with higher cardiogenic and therapeutic potential of the CDCs population. In addition, long-term BUT-treated CDCs had an increased ratio of collagen-III/collagen-I gene expression levels, and showed decreased release of inflammatory cytokines, overall supporting a less fibrosis-prone phenotype. In conclusion, β2 adrenergic receptor block positively affected the stemness vs commitment balance within CSs through the modulation of type1-EMT (so called "developmental"). These results further highlight type-1 EMT to be a key process affecting the features of resident cardiac progenitor cells, and mediating their response to the microenvironment.
Collapse
Affiliation(s)
- Francesca Pagano
- Department of Medical Surgical Sciences and Biotechnology, "La Sapienza" University of Rome, Italy
| | - Francesco Angelini
- Department of Medical Surgical Sciences and Biotechnology, "La Sapienza" University of Rome, Italy
| | - Camilla Siciliano
- Department of Medical Surgical Sciences and Biotechnology, "La Sapienza" University of Rome, Italy
| | - Julia Tasciotti
- Department of Medical Surgical Sciences and Biotechnology, "La Sapienza" University of Rome, Italy
| | - Giorgio Mangino
- Department of Medical Surgical Sciences and Biotechnology, "La Sapienza" University of Rome, Italy
| | - Elena De Falco
- Department of Medical Surgical Sciences and Biotechnology, "La Sapienza" University of Rome, Italy
| | - Roberto Carnevale
- Department of Medical Surgical Sciences and Biotechnology, "La Sapienza" University of Rome, Italy
| | - Sebastiano Sciarretta
- Department of Medical Surgical Sciences and Biotechnology, "La Sapienza" University of Rome, Italy; Department of AngioCardioNeurology, IRCCS Neuromed, Pozzilli, Italy
| | - Giacomo Frati
- Department of Medical Surgical Sciences and Biotechnology, "La Sapienza" University of Rome, Italy; Department of AngioCardioNeurology, IRCCS Neuromed, Pozzilli, Italy
| | - Isotta Chimenti
- Department of Medical Surgical Sciences and Biotechnology, "La Sapienza" University of Rome, Italy.
| |
Collapse
|
31
|
Wang T, Li Y, Lu HL, Meng QW, Cai L, Chen XS. β-Adrenergic Receptors : New Target in Breast Cancer. Asian Pac J Cancer Prev 2016; 16:8031-9. [PMID: 26745035 DOI: 10.7314/apjcp.2015.16.18.8031] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Preclinical studies have demonstrated that β-adrenergic receptor antagonists could improve the prognosis of breast cancer. However, the conclusions of clinical and pharmacoepidemiological studies have been inconsistent. This review was conducted to re-assess the relationship between beta-adrenoceptor blockers and breast cancer prognosis. MATERIALS AND METHODS The literature was searched from PubMed, EMBASE and Web of Nature (Thompson Reuters) databases through using key terms, such as breast cancer and beta- adrenoceptor blockers. RESULTS Ten publications met the inclusion criteria. Six suggested that receiving beta- adrenoceptor blockers reduced the risk of breast cancer-specific mortality, and three of them had statistical significance (hazard ratio (HR)=0.42; 95% CI=0.18-0.97; p=0.042). Two studies reported that risk of recurrence and distant metastasis (DM) were both significantly reduced. One study demonstrated that the risk of relapse- free survival (RFS) was raised significantly with beta-blockers (BBS) (HR= 0.30; 95% CI=0.10-0.87; p=0.027). One reported longer disease-free interval (Log Rank (LR)=6.658; p=0.011) in BBS users, but there was no significant association between overall survival (OS) and BBS (HR= 0.35; 95% CI=0.12-1.0; p=0.05) in five studies. CONCLUSIONS Through careful consideration, it is suggested that beta-adrenoceptor blockers use may be associated with improved prognosis in breast cancer patients. Nevertheless, larger size studies are needed to further explore the relationship between beta-blocker drug use and breast cancer prognosis.
Collapse
Affiliation(s)
- Ting Wang
- Department of Internal Medical Oncology, Harbin Medical University Cancer Hospital, Heilongjiang Province, China E-mail : ;
| | | | | | | | | | | |
Collapse
|
32
|
A natural food sweetener with anti-pancreatic cancer properties. Oncogenesis 2016; 5:e217. [PMID: 27065453 PMCID: PMC4848839 DOI: 10.1038/oncsis.2016.28] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 02/12/2016] [Accepted: 03/07/2016] [Indexed: 11/28/2022] Open
Abstract
Mogroside V is a triterpenoid isolated from the traditional Chinese medical plant Siraitia grosvenorii. Mogroside V has a high degree of sweetness and a low calorific content. Herein, we found that mogroside V possesses tumor growth inhibitory activity in in vitro and in vivo models of pancreatic cancer by promoting apoptosis and cell cycle arrest of pancreatic cancer cells (PANC-1 cells), which may in part be mediated through regulating the STAT3 signaling pathway. These results were confirmed in vivo in a mouse xenograft model of pancreatic cancer. In xenograft tumors, Ki-67 and PCNA, the most commonly used markers of tumor cell proliferation, were downregulated after intravenous administration of mogroside V. Terminal deoxynucleotidyl transferase dUTP nick end labeling assays showed that mogroside V treatment promoted apoptosis of pancreatic cancer cells in the xenograft tumors. Furthermore, we found that mogroside V treatment significantly reduced the expression of CD31-labeled blood vessels and of the pro-angiogenic factor vascular endothelial growth factor in the xenografts, indicating that mogroside V might limit the growth of pancreatic tumors by inhibiting angiogenesis and reducing vascular density. These results therefore demonstrate that the natural, sweet-tasting compound mogroside V can inhibit proliferation and survival of pancreatic cancer cells via targeting multiple biological targets.
Collapse
|
33
|
Choy C, Raytis JL, Smith DD, Duenas M, Neman J, Jandial R, Lew MW. Inhibition of β2-adrenergic receptor reduces triple-negative breast cancer brain metastases: The potential benefit of perioperative β-blockade. Oncol Rep 2016; 35:3135-42. [PMID: 27035124 PMCID: PMC4869944 DOI: 10.3892/or.2016.4710] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 11/05/2015] [Indexed: 01/02/2023] Open
Abstract
In response to recent studies, we investigated an association between perioperative β-blockade and breast cancer metastases. First, a retrospective study examining perioperative β-blocker use and cancer recurrence and metastases was conducted on 1,029 patients who underwent breast cancer surgery at the City of Hope Cancer Center between 2000 and 2010. We followed the clinical study and examined proliferation, migration, and invasion in vitro of primary and brain-metastatic breast cancer cells in response to β2-activation and inhibition. We also investigated in vivo the metastatic potential of propranolol-treated metastatic cells. For stage II breast cancer patients, perioperative β-blockade was associated with decreased cancer recurrence using Cox regression analysis (hazard's ratio =0.51; 95% CI: 0.23-0.97; p=0.041). Triple-negative (TN) brain-metastatic cells were found to have increased β2-adrenergic receptor mRNA and protein expression relative to TN primary cells. In response to β2-adrenergic receptor activation, TN brain-metastatic cells also exhibited increased cell proliferation and migration relative to the control. These effects were abrogated by propranolol. Propranolol decreased β2-adrenergic receptor-activated invasion. In vivo, propranolol treatment of TN brain-metastatic cells decreased establishment of brain metastases. Our results suggest that stress and corresponding β2-activation may promote the establishment of brain metastases of TN breast cancer cells. In addition, our data suggest a benefit to perioperative β-blockade during surgery-induced stress with respect to breast cancer recurrence and metastases.
Collapse
Affiliation(s)
- Cecilia Choy
- Division of Neurosurgery, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - John L Raytis
- Department of Anesthesiology, City of Hope, Duarte, CA 91010, USA
| | - David D Smith
- Department of Biostatistics, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Matthew Duenas
- Division of Neurosurgery, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Josh Neman
- Department of Neurosurgery, Keck School of Medicine at the University of Southern California, Los Angeles, CA 90089, USA
| | - Rahul Jandial
- Division of Neurosurgery, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Michael W Lew
- Department of Anesthesiology, City of Hope, Duarte, CA 91010, USA
| |
Collapse
|
34
|
Deutsch D, Deen S, Entschladen F, Coveney C, Rees R, Zänker KS, Powe DG. Alpha1B adrenoceptor expression is a marker of reduced survival and increased tumor recurrence in patients with endometrioid ovarian cancer. World J Obstet Gynecol 2016; 5:118-126. [DOI: 10.5317/wjog.v5.i1.118] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Revised: 08/27/2015] [Accepted: 11/11/2015] [Indexed: 02/05/2023] Open
Abstract
AIM: To investigate the expression patterns of different adrenoceptor isoforms in ovarian cancer and their association with survival and tumor recurrence.
METHODS: The protein expression levels of α1B, α2C and β2 adrenoceptor were assessed in unselected ovarian cancer using immunohistochemistry on microarrayed archival tissue samples. A database containing clinical and pathology parameters and follow-up was used to investigate the association between adrenoceptor isoform expression with ovarian specific survival and tumor recurrence, using univariate and multivariate statistical analysis.
RESULTS: Expression of α1B showed an association with reduced ovarian specific survival (P = 0.05; CI: 1.00-1.49) and increased tumor recurrence (P = 0.021, CI: 1.04-1.69) in the whole patient group. On sub-analysis the expression of α1B in endometrioid cancers (χ2 = 5.867, P = 0.015) was found to predict reduced ovarian specific survival and increased tumor recurrence independently of tumor grade, clinical stage and chemotherapy. An association with clinical outcome was not seen for α2C or β2 AR.
CONCLUSION: Alpha1B adrenoceptor protein was found to predict increased risk of tumor recurrence and reduced mortality in patients with endometrioid type ovarian cancer and should be investigated as a biomarker for identifying patients at increased risk of disease progression. Furthermore, α adrenergic receptor antagonists with α1B selectivity should be investigated as a possible adjuvant therapy for treating patients with endometrioid cancer. Proof of principle could be tested in a retrospective population study.
Collapse
|
35
|
Pon CK, Lane JR, Sloan EK, Halls ML. The β2-adrenoceptor activates a positive cAMP-calcium feedforward loop to drive breast cancer cell invasion. FASEB J 2015; 30:1144-54. [PMID: 26578688 DOI: 10.1096/fj.15-277798] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2015] [Accepted: 11/09/2015] [Indexed: 01/04/2023]
Abstract
Activation of the sympathetic nervous system by stress increases breast cancer metastasis in vivo. Preclinical studies suggest that stress activates β-adrenoceptors (βARs) to enhance metastasis from primary tumors and that β-blockers may be protective in breast cancer. However, the subtype of βAR that mediates this effect, as well as the signaling mechanisms underlying increased tumor cell dissemination, remain unclear. We show that the β2AR is the only functionally relevant βAR subtype in the highly metastatic human breast cancer cell line MDA-MB-231HM. β2AR activation results in elevated cAMP (formoterol pEC50 9.86 ± 0.32), increased intracellular Ca(2+) (formoterol pEC50 8.20 ± 0.33) and reduced phosphorylated ERK (pERK; formoterol pIC50 11.62 ± 0.31). We demonstrate that a highly amplified positive feedforward loop between the cAMP and Ca(2+) pathways is responsible for efficient inhibition of basal pERK. Importantly, activation of the β2AR increased invasion (formoterol area under the curve [AUC] relative to vehicle: 1.82 ± 0.36), which was dependent on the cAMP/Ca(2+) loop (formoterol AUC in the presence of 2'5'-dideoxyadenosine 0.64 ± 0.03, or BAPTA-AM 0.45 ± 0.23) but independent of inhibition of basal pERK1/2 (vehicle AUC with U0126 0.60 ± 0.30). Specifically targeting the positive feedforward cAMP/Ca(2+) loop may be beneficial for the development of therapeutics to slow disease progression in patients with breast cancer.
Collapse
Affiliation(s)
- Cindy K Pon
- *Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia; Cousins Center, UCLA Semel Institute for Neuroscience and Human Behavior, and Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, California USA; and Division of Cancer Surgery, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | - J Robert Lane
- *Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia; Cousins Center, UCLA Semel Institute for Neuroscience and Human Behavior, and Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, California USA; and Division of Cancer Surgery, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | - Erica K Sloan
- *Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia; Cousins Center, UCLA Semel Institute for Neuroscience and Human Behavior, and Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, California USA; and Division of Cancer Surgery, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | - Michelle L Halls
- *Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia; Cousins Center, UCLA Semel Institute for Neuroscience and Human Behavior, and Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, California USA; and Division of Cancer Surgery, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| |
Collapse
|
36
|
Gargiulo L, Copsel S, Rivero EM, Galés C, Sénard JM, Lüthy IA, Davio C, Bruzzone A. Differential β₂-adrenergic receptor expression defines the phenotype of non-tumorigenic and malignant human breast cell lines. Oncotarget 2015; 5:10058-69. [PMID: 25375203 PMCID: PMC4259405 DOI: 10.18632/oncotarget.2460] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Accepted: 09/06/2014] [Indexed: 11/27/2022] Open
Abstract
Breast cancer is the most frequent malignancy in women. Several reports demonstrated that adrenergic receptors (ARs) are involved in breast cancer. Here we observed that epinephrine (Epi), an endogenous AR agonist, caused opposite effects in non-tumorigenic (MCF-10A and HBL-100) and tumor cells (MCF-7 and MDA-MB-231). Thus, Epi, in non-tumor breast cells, as well as isoproterenol (β-agonist), in all cell lines, maintained a benign phenotype, decreasing cell proliferation and migration, and stimulating cell adhesion. β-AR expression and cAMP levels were higher in MCF-10A than in MCF-7 cells. β2-AR knock-down caused a significant increase of cell proliferation and migration, and a decrease of cell adhesion both in basal and in Iso-stimulated conditions. Coincidently, β2-AR over-expression induced a significant decrease of cell proliferation and migration, and an increase of cell adhesion. Therefore, β2-AR is implied in cell phenotype and its agonists or antagonists could eventually complement cancer therapy.
Collapse
Affiliation(s)
- Lucía Gargiulo
- Instituto de Biología y Medicina Experimental-CONICET, Vuelta de Obligado 2490, C1428ADN, CABA, Argentina
| | - Sabrina Copsel
- Instituto de Biología y Medicina Experimental-CONICET, Vuelta de Obligado 2490, C1428ADN, CABA, Argentina. Laboratorio de Farmacología de Receptores, Departamento de Farmacología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junin 956 (1113) CABA, Argentina
| | - Ezequiel M Rivero
- Instituto de Biología y Medicina Experimental-CONICET, Vuelta de Obligado 2490, C1428ADN, CABA, Argentina
| | - Céline Galés
- Institut des Maladies Métaboliques et Cardiovasculaires, Institut National de la Santé et de la Recherche Médicale, U1048, Université Toulouse III Paul Sabatier, F-31432 Toulouse, France
| | - Jean-Michel Sénard
- Institut des Maladies Métaboliques et Cardiovasculaires, Institut National de la Santé et de la Recherche Médicale, U1048, Université Toulouse III Paul Sabatier, F-31432 Toulouse, France
| | - Isabel A Lüthy
- Instituto de Biología y Medicina Experimental-CONICET, Vuelta de Obligado 2490, C1428ADN, CABA, Argentina
| | - Carlos Davio
- Laboratorio de Farmacología de Receptores, Departamento de Farmacología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junin 956 (1113) CABA, Argentina
| | - Ariana Bruzzone
- Instituto de Biología y Medicina Experimental-CONICET, Vuelta de Obligado 2490, C1428ADN, CABA, Argentina
| |
Collapse
|
37
|
Garona J, Pifano M, Orlando UD, Pastrian MB, Iannucci NB, Ortega HH, Podesta EJ, Gomez DE, Ripoll GV, Alonso DF. The novel desmopressin analogue [V4Q5]dDAVP inhibits angiogenesis, tumour growth and metastases in vasopressin type 2 receptor-expressing breast cancer models. Int J Oncol 2015; 46:2335-45. [PMID: 25846632 PMCID: PMC4441290 DOI: 10.3892/ijo.2015.2952] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 02/19/2015] [Indexed: 12/31/2022] Open
Abstract
Desmopressin (dDAVP) is a safe haemostatic agent with previously reported antitumour activity. It acts as a selective agonist for the V2 vasopressin membrane receptor (V2r) present on tumour cells and microvasculature. The purpose of this study was to evaluate the novel peptide derivative [V4Q5]dDAVP in V2r-expressing preclinical mouse models of breast cancer. We assessed antitumour effects of [V4Q5]dDAVP using human MCF-7 and MDA-MB-231 breast carcinoma cells, as well as the highly metastatic mouse F3II cell line. Effect on in vitro cancer cell growth was evaluated by cell proliferation and clonogenic assays. Cell cycle distribution was analysed by flow cytometry. In order to study the effect of intravenously administered [V4Q5]dDAVP on tumour growth and angiogenesis, breast cancer xenografts were generated in athymic mice. F3II cells were injected into syngeneic mice to evaluate the effect of [V4Q5]dDAVP on spontaneous and experimental metastatic spread. In vitro cytostatic effects of [V4Q5]dDAVP against breast cancer cells were greater than those of dDAVP, and associated with V2r-activated signal transduction and partial cell cycle arrest. In MDA-MB-231 xenografts, [V4Q5]dDAVP (0.3 μg/kg, thrice a week) reduced tumour growth and angiogenesis. Treatment of F3II mammary tumour-bearing immunocompetent mice resulted in complete inhibition of metastatic progression. [V4Q5]dDAVP also displayed greater antimetastatic efficacy than dDAVP on experimental lung colonisation by F3II cells. The novel analogue was well tolerated in preliminary acute toxicology studies, at doses ≥300-fold above that required for anti-angiogenic/antimetastatic effects. Our data establish the preclinical activity of [V4Q5]dDAVP in aggressive breast cancer, providing the rationale for further clinical trials.
Collapse
Affiliation(s)
- Juan Garona
- Laboratory of Molecular Oncology, National University of Quilmes, Bernal, B1876BXD, Buenos Aires, Argentina
| | - Marina Pifano
- Laboratory of Molecular Oncology, National University of Quilmes, Bernal, B1876BXD, Buenos Aires, Argentina
| | - Ulises D Orlando
- Biomedical Research Institute (INBIOMED), Department of Human Biochemistry, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Maria B Pastrian
- School of Pharmacy and Biochemistry, University of Buenos Aires, Buenos Aires, Argentina
| | - Nancy B Iannucci
- School of Pharmacy and Biochemistry, University of Buenos Aires, Buenos Aires, Argentina
| | - Hugo H Ortega
- Institute of Veterinary Sciences (ICIVET-CONICET), National University of Litoral, Esperanza, Santa Fe, Argentina
| | - Ernesto J Podesta
- Biomedical Research Institute (INBIOMED), Department of Human Biochemistry, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Daniel E Gomez
- Laboratory of Molecular Oncology, National University of Quilmes, Bernal, B1876BXD, Buenos Aires, Argentina
| | - Giselle V Ripoll
- Laboratory of Molecular Oncology, National University of Quilmes, Bernal, B1876BXD, Buenos Aires, Argentina
| | - Daniel F Alonso
- Laboratory of Molecular Oncology, National University of Quilmes, Bernal, B1876BXD, Buenos Aires, Argentina
| |
Collapse
|
38
|
Leung HWC, Hung LL, Chan ALF, Mou CH. Long-Term Use of Antihypertensive Agents and Risk of Breast Cancer: A Population-Based Case-Control Study. Cardiol Ther 2015; 4:65-76. [PMID: 25657096 PMCID: PMC4472646 DOI: 10.1007/s40119-015-0035-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Indexed: 01/02/2023] Open
Abstract
INTRODUCTION To evaluate the risk of breast cancer associated with long-term use of antihypertensive agents (AHs) in Taiwanese women with hypertension. METHODS A search of the Taiwan National Health Insurance Research Database identified 330,699 patients with hypertension who were treated with antihypertensive drugs between January 1, 1998 and December 31, 2011. Logistic regression models were used to estimate the odds ratios (ORs) and 95% confidence intervals (CIs) between the use of AHs and breast cancer risk, adjusted for other types of antihypertensive drugs, statins and co-morbidities. RESULTS Among the AHs used to treat the hypertensive women enrolled in our study, our analysis revealed that those treated with one specific particular class of beta-blockers (beta-1 selective beta-blockers) had an increased risk for breast cancer. We also found that the ever-use of calcium channel blockers (CCBs; i.e. for 13 years) was associated with breast cancer in an adjusted model (OR 1.09; 95% CI 1.03-1.16). However, the use of nonselective beta-blockers, selective and nonselective alpha-blockers, angiotensin-converting enzyme inhibitors and angiotensin II antagonists were not associated with breast cancer risk. CONCLUSION Based on the results of our analysis, long-term use of CCBs or beta-1 selective beta-blockers are likely to be associated with the risk of breast cancer. Further large comprehensive population-based studies to support our findings are required for confirmation of this conclusion.
Collapse
Affiliation(s)
- Henry W C Leung
- Department of Radiation Oncology, China Medical University-An Nan Hospital, Tainan, Taiwan
| | | | | | | |
Collapse
|
39
|
Abdin AA, Soliman NA, Saied EM. Effect of propranolol on IL-10, visfatin, Hsp70, iNOS, TLR2, and survivin in amelioration of tumor progression and survival in Solid Ehrlich Carcinoma-bearing mice. Pharmacol Rep 2014; 66:1114-21. [PMID: 25443743 DOI: 10.1016/j.pharep.2014.07.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Revised: 07/21/2014] [Accepted: 07/23/2014] [Indexed: 01/25/2023]
Abstract
BACKGROUND β-Adrenergic signaling could contribute to initiation and progression of breast cancer. This research investigated some potential mechanisms of propranolol in amelioration of progression and survival in breast cancer. METHODS AND RESULTS Solid Ehrlich Carcinoma (SEC) xenograft model was induced in 30 mice divided into 3 groups; where group I served as untreated SEC group. In groups II and III, propranolol treatment i.p. in low (5mg/kg) and high dose (10mg/kg) caused significant increase in interleukin-10 (IL-10) and decrease in heat shock protein 70 (Hsp70) and inducible nitric oxide synthase (iNOS) activity with non significant change in visfatin in tumor tissues compared to untreated SEC. In untreated SEC, tumor volume (V) exhibited significant negative correlation with IL-10 levels and toll like receptor 2 (TLR2) expression with significant positive correlation with Hsp70 levels and iNOS activity. While propranolol in either doses caused reduction of tumor volume (V), and improved percentage tumor growth inhibition (% TGI) only its high dose exhibited significant impact on survival rate. Propranolol dose-dependent effect was evident for IL-10 and Hsp70, and even only the high dose significantly increased and decreased TLR2 and survivin, respectively. This comes in favor of recommending high dose of propranolol in cancer therapy. Nonetheless, use of low dose cannot be ignored when benefit to risk balance have to be considered. CONCLUSIONS Propranolol could provide palliative effects in progression and survival of breast cancer that are mainly mediated via direct immunomodulatory and apoptotic mechanisms and probably associated with indirect anti-angiogenic activity.
Collapse
Affiliation(s)
- Amany A Abdin
- Department of Pharmacology, Faculty of Medicine, Tanta University, Tanta, Egypt.
| | - Nema A Soliman
- Department of Medical Biochemistry, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Eman M Saied
- Department of Pathology, Faculty of Medicine, Kafr El-Sheikh University, Tanta, Egypt
| |
Collapse
|
40
|
Bruzzone A, Saulière A, Finana F, Sénard JM, Lüthy I, Galés C. Dosage-dependent regulation of cell proliferation and adhesion through dual β2-adrenergic receptor/cAMP signals. FASEB J 2013; 28:1342-54. [PMID: 24308976 DOI: 10.1096/fj.13-239285] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The role of β-adrenergic receptors (β-ARs) remains controversial in normal and tumor breast. Herein we explore the cAMP signaling involved in β-AR-dependent control of proliferation and adhesion of nontumor human breast cell line MCF-10A. Low concentrations of a β-agonist, isoproterenol (ISO), promote cell adhesion (87.5% cells remaining adherent to the plastic dishes following specific detachment vs. 35.0% in control, P<0.001), while increasing concentrations further engages an additional 36% inhibition of Erk1/2 phosphorylation (p-Erk1/2)-dependent cell proliferation (P<0.01). Isoproterenol dose response on cell adhesion was fitted to a 2-site curve (EC50(1): 16.5±11.5 fM, EC50(2): 4.08±3.09 nM), while ISO significantly inhibited p-Erk1/2 according to a 1-site model (EC50: 0.25±0.13 nM). Using β-AR-selective agonist/antagonists and cAMP analogs/inhibitors, we identified a dosage-dependent signaling in which low ISO concentrations target a β2-AR population localized in raft microdomains and stimulate a Gs/cAMP/Epac/adhesion-signaling module, while higher concentrations engage a concomitant activation of another β2-AR population outside rafts and inhibit the proliferation by a Gs/cAMP/PKA-dependent signaling module. Our data provide a new molecular basis for the dose-dependent switch of β-AR signaling. This study also sheds light on a new cAMP pathway core mechanism with a single receptor triggering dual cAMP signaling controlled by PKA or Epac but with different cellular outputs.
Collapse
Affiliation(s)
- Ariana Bruzzone
- 2Inserm U1048, Institut des Maladies Métaboliques et Cardiovasculaires, I2MC, 1, avenue Jean-Poulhès, BP84225, 31432 Toulouse Cedex 4, France.
| | | | | | | | | | | |
Collapse
|
41
|
β-Adrenergic system, a backstage manipulator regulating tumour progression and drug target in cancer therapy. Semin Cancer Biol 2013; 23:533-42. [PMID: 24012659 DOI: 10.1016/j.semcancer.2013.08.009] [Citation(s) in RCA: 125] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Accepted: 08/27/2013] [Indexed: 12/18/2022]
Abstract
β-Adrenoceptors are broadly distributed in various tissues of the body. Stress hormones regulate a panel of important physiological functions and disease states including cancer. Nicotine and its derivatives could stimulate the release of stress hormones from cancer cells, leading to the promotion of cancer development. β-Blockers have been widely used to control hypertension for decades. Recently, these agents could have significant implications in cancer therapy through blockade of adrenoceptors in tumour tissues. In this review, we summarize recent advancements about the influence of stress hormones, nicotine and β-adrenoceptors on cancer cell proliferation, apoptosis, invasion and metastasis, and also tumour vasculature normalization. Relevant signal pathways and potential value of β-blockers in the treatment of cancer are also discussed in this review.
Collapse
|
42
|
Kafetzopoulou LE, Boocock DJ, Dhondalay GKR, Powe DG, Ball GR. Biomarker identification in breast cancer: Beta-adrenergic receptor signaling and pathways to therapeutic response. Comput Struct Biotechnol J 2013; 6:e201303003. [PMID: 24688711 PMCID: PMC3962150 DOI: 10.5936/csbj.201303003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Revised: 03/19/2013] [Accepted: 03/21/2013] [Indexed: 12/20/2022] Open
Abstract
Recent preclinical studies have associated beta-adrenergic receptor (β-AR) signaling with breast cancer pathways such as progression and metastasis. These findings have been supported by clinical and epidemiological studies which examined the effect of beta-blocker therapy on breast cancer metastasis, recurrence and mortality. Results from these studies have provided initial evidence for the inhibition of cell migration in breast cancer by beta-blockers and have introduced the beta-adrenergic receptor pathways as a target for therapy. This paper analyzes gene expression profiles in breast cancer patients, utilising Artificial Neural Networks (ANNs) to identify molecular signatures corresponding to possible disease management pathways and biomarker treatment strategies associated with beta-2-adrenergic receptor (ADRB2) cell signaling. The adrenergic receptor relationship to cancer is investigated in order to validate the results of recent studies that suggest the use of beta-blockers for breast cancer therapy. A panel of genes is identified which has previously been reported to play an important role in cancer and also to be involved in the beta-adrenergic receptor signaling.
Collapse
Affiliation(s)
- Liana E Kafetzopoulou
- The John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, NG11 8NS, UK
| | - David J Boocock
- The John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, NG11 8NS, UK
| | - Gopal Krishna R Dhondalay
- The John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, NG11 8NS, UK
| | - Desmond G Powe
- The John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, NG11 8NS, UK ; Department of Cellular Pathology, Nottingham University Hospitals Trust and University of Nottingham, Nottingham, NG7 2UH, UK
| | - Graham R Ball
- The John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, NG11 8NS, UK
| |
Collapse
|
43
|
Abstract
β-adrenergic signaling modulates key signaling pathways that are important for tumor-promoting processes, and numerous mechanisms of action have been elucidated. Preclinical studies have demonstrated that β-adrenergic antagonists, or β-blockers, can block multiple fundamental biologic processes underlying the progression and metastasis of tumors, including the inhibition of cell proliferation, migration, invasion, resistance to programmed cell death, and tumor angiogenesis and metastasis. Human pharmacoepidemiologic studies suggest that β-blockers have a role in inhibiting cancer progression and metastasis in combination with standard therapies. Furthermore, a number of prospective studies have demonstrated that β-blockers are effective at halting infantile hemangioma growth. These findings shed light on the novel perspective of using β-blockers as a class of potential antitumor agents in clinical oncology.
Collapse
Affiliation(s)
- Yi Ji
- Division of Oncology, Department of Pediatric Surgery, Children's Hospital of Fudan University, Shanghai, China
| | | | | | | | | |
Collapse
|
44
|
Paul RK, Ramamoorthy A, Scheers J, Wersto RP, Toll L, Jimenez L, Bernier M, Wainer IW. Cannabinoid receptor activation correlates with the proapoptotic action of the β2-adrenergic agonist (R,R')-4-methoxy-1-naphthylfenoterol in HepG2 hepatocarcinoma cells. J Pharmacol Exp Ther 2012; 343:157-66. [PMID: 22776956 PMCID: PMC3464034 DOI: 10.1124/jpet.112.195206] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Accepted: 07/06/2012] [Indexed: 12/17/2022] Open
Abstract
Inhibition of cell proliferation by fenoterol and fenoterol derivatives in 1321N1 astrocytoma cells is consistent with β(2)-adrenergic receptor (β(2)-AR) stimulation. However, the events that result in fenoterol-mediated control of cell proliferation in other cell types are not clear. Here, we compare the effect of the β(2)-AR agonists (R,R')-fenoterol (Fen) and (R,R')-4-methoxy-1-naphthylfenoterol (MNF) on signaling and cell proliferation in HepG2 hepatocarcinoma cells by using Western blotting and [(3)H]thymidine incorporation assays. Despite the expression of β(2)-AR, no cAMP accumulation was observed when cells were stimulated with isoproterenol or Fen, although the treatment elicited both mitogen-activated protein kinase and phosphatidylinositol 3-kinase/Akt activation. Unexpectedly, isoproterenol and Fen promoted HepG2 cell growth, but MNF reduced proliferation together with increased apoptosis. The mitogenic responses of Fen were attenuated by 3-(isopropylamino)-1-[(7-methyl-4-indanyl)oxy]butan-2-ol (ICI 118,551), a β(2)-AR antagonist, whereas those of MNF were unaffected. Because of the coexpression of β(2)-AR and cannabinoid receptors (CBRs) and their impact on HepG2 cell proliferation, these Gα(i)/Gα(o)-linked receptors may be implicated in MNF signaling. Cell treatment with (R)-(+)-[2,3-dihydro-5-methyl-3-(4-morpholinylmethyl)pyrrolo[1,2,3-de]-1,4-benzoxazin-6-yl]-1-napthalenylmethanone (WIN 55,212-2), a synthetic agonist of CB(1)R and CB(2)R, led to growth inhibition, whereas inverse agonists of these receptors blocked MNF mitogenic responses without affecting Fen signaling. MNF responses were sensitive to pertussis toxin. The β(2)-AR-deficient U87MG cells were refractory to Fen, but responsive to the antiproliferative actions of MNF and WIN 55,212-2. The data indicate that the presence of the naphthyl moiety in MNF results in functional coupling to the CBR pathway, providing one of the first examples of a dually acting β(2)-AR-CBR ligand.
Collapse
Affiliation(s)
- Rajib K Paul
- Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224-6825, USA
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Pérez Piñero C, Bruzzone A, Sarappa MG, Castillo LF, Lüthy IA. Involvement of α2- and β2-adrenoceptors on breast cancer cell proliferation and tumour growth regulation. Br J Pharmacol 2012; 166:721-36. [PMID: 22122228 DOI: 10.1111/j.1476-5381.2011.01791.x] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND AND PURPOSE β-Adrenoceptors are expressed in human and experimental animal breast cancer cells. However, the effect of the agonists and antagonists reported on cell proliferation and tumour growth was paradoxical, precluding their utilization as possible adjuvant therapy, mainly in the cases of refractory tumours. EXPERIMENTAL APPROACH β-Adrenoceptor expression was analysed by immunofluorescence and RT-PCR. Cell proliferation was assessed by [(3) H]-thymidine incorporation, tumour growth by measuring with a calliper and ERK 1/2 phosphorylation by Western blotting. KEY RESULTS β(2) -Adrenoceptor expression was confirmed in the mouse and human cells tested. Cell proliferation was increased by adrenaline (by α(2) -adrenoceptor action) and decreased in every tested cell line by the β-adrenoceptor agonist isoprenaline and the β(2) -adrenoceptor agonist salbutamol. Isoprenaline and salbutamol reduced tumour growth in every tumour tested (mouse C4-HD and CC4-3-HI and human IBH-4, IBH-6 and MDA-MB-231 cell lines growing as xenografts in nude mice). These effects were reversed by the β-adrenoceptor antagonist propranolol. The α(2) -adrenoceptor antagonist rauwolscine and the β(2) -adrenoceptor agonist salbutamol were equally effective in diminishing tumour growth. ERK 1/2 activation analysed in IBH-4 tumours correlated with tumour growth, with the β-adrenoceptor agonists decreasing its activation. Inhibition of ERK 1/2 phosphorylation in vitro was mainly mediated by the PKA pathway. CONCLUSIONS AND IMPLICATIONS In our experimental models, the β-adrenoceptor agonists inhibited breast cancer cell proliferation and tumour growth, probably mediated by inhibition of ERK 1/2 phosphorylation. The β-adrenoceptor agonists were as effective as the α(2) -adrenoceptor antagonist rauwolscine, providing possible novel adjuvant treatments for breast cancer.
Collapse
Affiliation(s)
- C Pérez Piñero
- Instituto de Biología y Medicina Experimental - CONICET, Vuelta de Obligado 2490, C1428ADN Ciudad Autónoma de Buenos Aires, Argentina
| | | | | | | | | |
Collapse
|
46
|
Barron TI, Sharp L, Visvanathan K. Beta-adrenergic blocking drugs in breast cancer: a perspective review. Ther Adv Med Oncol 2012; 4:113-25. [PMID: 22590485 DOI: 10.1177/1758834012439738] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
The purpose of this review is to present the preclinical, epidemiological and clinical data relevant to the association between β-blockers and breast cancer progression. Preclinical studies have shown that β-adrenergic receptor (β-AR) signalling can inhibit multiple cellular processes involved in breast cancer progression and metastasis, including extracellular matrix invasion, expression of inflammatory and chemotactic cytokines, angiogenesis and tumour immune responses. This has led to the hypothesis that the commonly prescribed class of β-AR antagonist drugs (β-blockers) may favourably impact cancer progression. A number of recent pharmacoepidemiological studies have examined the association between β-blocker exposure and breast cancer progression. The results from these studies have suggested a potential role for targeting the β-AR pathway in breast cancer patients. Larger observational studies are, however, required to confirm these results. Questions regarding the type of β-blocker, predictive biomarkers or tumour characteristics, appropriate treatment paradigms and, most importantly, efficacy must also be answered in randomized clinical studies before β-blockers can be considered a therapeutic option for patients with breast cancer.
Collapse
Affiliation(s)
- Thomas I Barron
- Department of Pharmacology & Therapeutics, Trinity Centre for Health Sciences, St James's Hospital, Dublin 8, Ireland
| | | | | |
Collapse
|
47
|
Barron TI, Sharp L, Visvanathan K. Beta-adrenergic blocking drugs in breast cancer: a perspective review. Ther Adv Med Oncol 2012. [PMID: 22590485 DOI: 10.1177/1758834012439738,10.1177_1758834012439738] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The purpose of this review is to present the preclinical, epidemiological and clinical data relevant to the association between β-blockers and breast cancer progression. Preclinical studies have shown that β-adrenergic receptor (β-AR) signalling can inhibit multiple cellular processes involved in breast cancer progression and metastasis, including extracellular matrix invasion, expression of inflammatory and chemotactic cytokines, angiogenesis and tumour immune responses. This has led to the hypothesis that the commonly prescribed class of β-AR antagonist drugs (β-blockers) may favourably impact cancer progression. A number of recent pharmacoepidemiological studies have examined the association between β-blocker exposure and breast cancer progression. The results from these studies have suggested a potential role for targeting the β-AR pathway in breast cancer patients. Larger observational studies are, however, required to confirm these results. Questions regarding the type of β-blocker, predictive biomarkers or tumour characteristics, appropriate treatment paradigms and, most importantly, efficacy must also be answered in randomized clinical studies before β-blockers can be considered a therapeutic option for patients with breast cancer.
Collapse
Affiliation(s)
- Thomas I Barron
- Department of Pharmacology & Therapeutics, Trinity Centre for Health Sciences, St James's Hospital, Dublin 8, Ireland
| | | | | |
Collapse
|
48
|
Insel PA, Zhang L, Murray F, Yokouchi H, Zambon AC. Cyclic AMP is both a pro-apoptotic and anti-apoptotic second messenger. Acta Physiol (Oxf) 2012; 204:277-87. [PMID: 21385327 DOI: 10.1111/j.1748-1716.2011.02273.x] [Citation(s) in RCA: 168] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The second messenger cyclic AMP (cAMP) can either stimulate or inhibit programmed cell death (apoptosis). Here, we review examples of cell types that show pro-apoptotic or anti-apoptotic responses to increases in cAMP. We also show that cells can have both such responses, although predominantly having one or the other. Protein kinase A (PKA)-promoted changes in phosphorylation and gene expression can mediate pro-apoptotic responses, such as in murine S49 lymphoma cells, based on evidence that mutants lacking PKA fail to undergo cAMP-promoted, mitochondria-dependent apoptosis. Mechanisms for the anti-apoptotic response to cAMP likely involve Epac (Exchange protein activated by cAMP), a cAMP-regulated effector that is a guanine nucleotide exchange factor (GEF) for the low molecular weight G-protein, Rap1. Therapeutic approaches that activate PKA-mediated pro-apoptosis or block Epac-mediated anti-apoptotisis may provide a means to enhance cell killing, such as in certain cancers. In contrast, efforts to block PKA or stimulate Epac have the potential to be useful in diseases settings (such as heart failure) associated with cAMP-promoted apoptosis.
Collapse
Affiliation(s)
- P A Insel
- Department of Pharmacology, University of California, San Diego, La Jolla, 92093-0636, USA.
| | | | | | | | | |
Collapse
|
49
|
Mi D, Zhang M, Yan JD, Zhang J, Wang X, Wang Q, Yang S, Zhu TH. PTHrP inhibits BMP-6 expression through the PKA signaling pathway in breast cancer cells. J Cancer Res Clin Oncol 2011; 137:295-303. [PMID: 20401668 DOI: 10.1007/s00432-010-0883-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2010] [Accepted: 03/22/2010] [Indexed: 10/19/2022]
Abstract
BACKGROUND PTHrP, a mediator of humoral hypercalcemia of malignancy, is considered as a potential activator to induce breast cancer cells metastasizing to bone. However, recent clinical evidences and basal research results prove that PTHrP expression in primary tumors indicates good prognosis. BMP-6, as a member of TGF-β superfamily, is closely correlated with tumor differentiation and skeletal metastasis. PURPOSE These experiments were designed to investigate the molecular mechanism of PTHrP regulating BMP-6 in breast cancer cells. METHODS AND RESULTS Through detecting mRNA expression levels of PTHrP and BMP-6 in 35 breast cancer specimens, the two genes' expression were proved to be negatively correlated. Moreover, PTHrP (1-40), instead of PTHrP (107-139), inhibited BMP-6 mRNA expression in MCF-7 cells, indicating that PTHrP exerts its effect on BMP-6 through membranous PTHrP receptor. Inhibitors against signaling pathways downstream of PTHrP were utilized. H89, the PKA pathway inhibitor, eliminated the inhibitory effect of PTHrP on BMP-6. In addition, silencing of BMP-6 strengthened the antimitogenic effect of PTHrP. CONCLUSIONS These results suggest that PTHrP acts as the upstream molecule of BMP-6, and exerts antimitogenic effect via reducing BMP-6 mRNA expression through PKA signaling pathway in breast cancer cells.
Collapse
Affiliation(s)
- Dong Mi
- Laboratory of Molecular Genetics, Medical College, Nankai University, Tianjin 300071, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Toll L, Jimenez L, Waleh N, Jozwiak K, Woo AYH, Xiao RP, Bernier M, Wainer IW. {Beta}2-adrenergic receptor agonists inhibit the proliferation of 1321N1 astrocytoma cells. J Pharmacol Exp Ther 2011; 336:524-32. [PMID: 21071556 PMCID: PMC3033720 DOI: 10.1124/jpet.110.173971] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2010] [Accepted: 11/01/2010] [Indexed: 12/27/2022] Open
Abstract
Astrocytomas and glioblastomas have been particularly difficult to treat and refractory to chemotherapy. However, significant evidence has been presented that demonstrates a decrease in astrocytoma cell proliferation subsequent to an increase in cAMP levels. The 1321N1 astrocytoma cell line, as well as other astrocytomas and glioblastomas, expresses β(2)-adrenergic receptors (β(2)-ARs) that are coupled to G(s) activation and consequent cAMP production. Experiments were conducted to determine whether the β(2)-AR agonist (R,R')-fenoterol and other β(2)-AR agonists could attenuate mitogenesis and, if so, by what mechanism. Receptor binding studies were conducted to characterize β(2)-AR found in 1321N1 and U118 cell membranes. In addition, cells were incubated with (R,R')-fenoterol and analogs to determine their ability to stimulate intracellular cAMP accumulation and inhibit [(3)H]thymidine incorporation into the cells. 1321N1 cells contain significant levels of β(2)-AR as determined by receptor binding. (R,R')-fenoterol and other β(2)-AR agonists, as well as forskolin, stimulated cAMP accumulation in a dose-dependent manner. Accumulation of cAMP induced a decrease in [(3)H]thymidine incorporation. There was a correlation between concentration required to stimulate cAMP accumulation and inhibit [(3)H]thymidine incorporation. U118 cells have a reduced number of β(2)-ARs and a concomitant reduction in the ability of β(2)-AR agonists to inhibit cell proliferation. These studies demonstrate the efficacy of β(2)-AR agonists for inhibition of growth of the astrocytoma cell lines. Because a significant portion of brain tumors contain β(2)-ARs to a greater extent than whole brain, (R,R')-fenoterol, or some analog, may be useful in the treatment of brain tumors after biopsy to determine β(2)-AR expression.
Collapse
Affiliation(s)
- L Toll
- SRI International, Menlo Park, California, USA
| | | | | | | | | | | | | | | |
Collapse
|