1
|
Zhang R, Chen M, Deng Z, Kong L, Shen B, Zhang L. Delta Opioid Peptide Targets Brain Microvascular Endothelial Cells Reducing Apoptosis to Relieve Hypoxia-Ischemic/Reperfusion Injury. Pharmaceutics 2022; 15:pharmaceutics15010046. [PMID: 36678674 PMCID: PMC9861451 DOI: 10.3390/pharmaceutics15010046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 12/15/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
Stroke is one of the leading causes of death. (D-ala2, D-leu5) enkephalin (DADLE) is a synthetic peptide and highly selective delta opioid receptor (δOR) agonist that has exhibited protective properties in ischemia. However, the specific target and mechanism are still unclear. The present study explores the expression of δOR on brain microvascular endothelial cells (BMECs) and whether DADLE could relieve I/R-induced injury by reducing apoptosis. A lateral ventricular injection of DADLE for pretreatment, the neurofunctional behavior score, and TTC staining, were used to evaluate the protective effect of DADLE. Immunofluorescence technology was used to label different types of cells with apoptosis-positive signals to test co-localization status. Primary cultured BMECs were separated and treated with DADLE, accompanied by OGD/R. The CCK-8 test was conducted to evaluate cell viability and TdT-mediated dUTP Nick-end Labelling (TUNEL) staining to test apoptosis levels. The levels of apoptosis-related proteins were analyzed by Western blotting. The co-localization results showed that BMECs, but not astrocytes, microglia, or neurons, presented mostly TUNEL-positive signals, especially in the Dentate gyrus (DG) area of the hippocampus. Either activation of δORs on rats' brains or primary BMECs mainly reduce cellular apoptosis and relieve the injury. Interference with the expression δOR could block this effect. DADLE also significantly increased levels of Bcl-2 and reduced levels of Bax. δOR's expressions can be detected on the BMECs, but not on the HEK293 cells, by Western blotting and IFC. Therefore, DADLE exerts a cytoprotective effect, primarily under hypoxia-ischemic injury/reperfusion conditions, by targeting BMECs to inhibit apoptosis.
Collapse
Affiliation(s)
- Ran Zhang
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Meixuan Chen
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Zhongfang Deng
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Lingchao Kong
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei 230032, China
| | - Bing Shen
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Lesha Zhang
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
- Correspondence:
| |
Collapse
|
2
|
Giakomidi D, Khemiri S, Mahbuba W, McVey DG, Al-Janabi F, Guerrini R, Calo G, Ye S, Lambert DG. Nociceptin/Orphanin FQ receptor expression in primary human umbilical vein endothelial cells is not regulated by exposure to breast cancer cell media or angiogenic stimuli. BJA OPEN 2022; 4:100110. [PMID: 37588788 PMCID: PMC10430811 DOI: 10.1016/j.bjao.2022.100110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 10/20/2022] [Indexed: 08/18/2023]
Abstract
Background Opioid receptors are naloxone-sensitive (MOP [mu: μ], DOP [delta: δ], and KOP [kappa: κ]) and naloxone-insensitive Nociceptin/Orphanin FQ (N/OFQ) peptide receptor (NOP). Clinically, most opioid analgesics target MOP. Angiogenesis is the formation of new blood vessels and involves endothelial cell activation, proliferation, and migration. The effect of opioids on this process is controversial with no data for NOP receptor ligands. Methods We used patient-derived human umbilical vein endothelial cells (HUVECs) treated with media from the Michigan Cancer Foundation-7 (MCF-7) breast cancer cells or vascular endothelial growth factor (VEGF; 10 ng ml-1) and fibroblast growth factor (FGF; 10 ng ml-1) as angiogenic stimuli. We have measured (i) NOP/MOP messenger RNA, (ii) receptor protein using N/OFQATTO594 and DermorphinATTO488 as fluorescent probes for NOP and MOP, and (iii) NOP/MOP function in a wound healing assay (crude measure of migration that occurs during angiogenesis). Results HUVEC lines from 32 patients were used. Using all 32 lines, mRNA for NOP but not MOP was detected. This was unaffected by media from MCF-7 cells or VEGF/FGF. There was no binding of either N/OFQATTO594(NOP) or DermorphinATTO488(MOP) in the absence or presence of angiogenic stimuli (six lines tested). In the absence of MOP mRNA, this was expected. Whilst MCF-7 conditioned medium (not VEGF/FGF) reduced wound healing per se (14 lines tested), there was no effect of N/OFQ (NOP ligand) or morphine (MOP ligand). Conclusions Media from MCF-7 breast cancer cells or VEGF/FGF as angiogenic stimuli did not influence NOP translation into receptor protein. MOP was absent. In the absence of constitutive or inducible MOP/NOP, there was no effect on wound healing as a measure of angiogenesis.
Collapse
Affiliation(s)
- Despina Giakomidi
- Department of Cardiovascular Sciences, Anaesthesia, Critical Care and Pain Management, Hodgkin Building, University of Leicester, Leicester, UK
| | - Sonja Khemiri
- Department of Cardiovascular Sciences, Anaesthesia, Critical Care and Pain Management, Hodgkin Building, University of Leicester, Leicester, UK
| | - Wadhah Mahbuba
- Department of Cardiovascular Sciences, Anaesthesia, Critical Care and Pain Management, Hodgkin Building, University of Leicester, Leicester, UK
| | - David G. McVey
- Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Centre, University of Leicester, Leicester, UK
| | - Fatin Al-Janabi
- Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Centre, University of Leicester, Leicester, UK
| | - Remo Guerrini
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Ferrara, Italy
| | - Girolamo Calo
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Shu Ye
- Department of Cardiovascular Sciences and NIHR Leicester Biomedical Research Centre, University of Leicester, Leicester, UK
| | - David G. Lambert
- Department of Cardiovascular Sciences, Anaesthesia, Critical Care and Pain Management, Hodgkin Building, University of Leicester, Leicester, UK
| |
Collapse
|
3
|
Rezaeiamiri E, Asadi M, Hosseini FS, Amanlou A, Dehpour AR, Amanlou M. Thebaine Derivatives as a New Regulator of Tumor Angiogenesis. Polycycl Aromat Compd 2022. [DOI: 10.1080/10406638.2021.1922471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Elnaz Rezaeiamiri
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehdi Asadi
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Faezeh Sadat Hosseini
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Arash Amanlou
- Faculty of Specialized Veterinary Sciences, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Ahmad Reza Dehpour
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Massoud Amanlou
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
4
|
Abstract
Current experimental stroke research has evolved to focus on detailed understanding of the brain’s self-protective and restorative mechanisms, and harness this knowledge for development of new therapies. In this context, the role of peptidases and neuropeptides is of growing interest. In this focused review, peptidase neurolysin (Nln) and its extracellular peptide substrates are briefly discussed in relation to pathophysiology of ischemic stroke. Upregulation of Nln following stroke is viewed as a compensatory cerebroprotective mechanism in the acute phase of stroke, because the main neuropeptides inactivated by Nln are neuro/cerebrotoxic (bradykinin, substance P, neurotensin, angiotensin II, hemopressin), whereas the peptides generated by Nln are neuro/cerebroprotective (angiotensin-(1–7), Leu-/Met-enkephalins). This notion is confirmed by experimental studies documenting aggravation of stroke outcomes in mice after inhibition of Nln following stroke, and dramatic improvement of stroke outcomes in mice overexpressing Nln in the brain. The role of Nln in the (sub)chronic phase of stroke is less clear and it is likely, that this peptidase does not have a major role in neural repair mechanisms. This is because, the substrates of Nln are less uniform in modulating neurorestorative mechanisms in one direction, some appearing to have neural repair enhancing/stimulating potential, whereas others doing the opposite. Future studies focusing on the role of Nln in pathophysiology of stroke should determine its potential as a cerebroprotective target for stroke therapy, because its unique ability to modulate multiple neuropeptide systems critically involved in brain injury mechanisms is likely advantageous over modulation of one pathogenic pathway for stroke pharmacotherapy.
Collapse
Affiliation(s)
- Vardan T Karamyan
- Department of Pharmaceutical Sciences and Center for Blood Brain Barrier Research, School of Pharmacy, TTUHSC, Amarillo, TX, USA
| |
Collapse
|
5
|
Opioid receptors beyond pain control: The role in cancer pathology and the debated importance of their pharmacological modulation. Pharmacol Res 2020; 159:104938. [DOI: 10.1016/j.phrs.2020.104938] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 03/24/2020] [Accepted: 05/15/2020] [Indexed: 12/15/2022]
|
6
|
Gein SV, Baeva TA. [Endomorphins: structure, localization, immunoregulatory activity]. ACTA ACUST UNITED AC 2020; 66:78-86. [PMID: 33351316 DOI: 10.14341/probl10364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 12/09/2019] [Accepted: 01/25/2020] [Indexed: 11/06/2022]
Abstract
Endomorphins – endogenous tetrapeptides with the highest affinity for the µ-opioid receptor. Currently, two tetrapeptides that differ in one amino acid residue have been isolated and characterized. The structure of endomorphins differs from the structure of members of three main families of opioid peptides: endorphins, enkephalins, and dynorphins, which contain the same N-terminal sequence. In the central nervous system, endomorphins are distributed everywhere, where they are primarily responsible for antinociception. Distribution of endomorphins in the immune system, similar to that of other opioid peptides, has allowed to suggest their active participation in the processes of immune regulation. This review summarizes modern views on the structure of endomorphins, their localization, possible intracellular mechanisms of signal transmission and their effects on the processes of activation, proliferation and differentiation of cells of innate and adaptive immunity. Endomorphins actively modulate the functions of the cells of the immune system. Peptides predominantly suppress adaptive immunity reactions. There effects on the functions of innate immunity cells (granulocytes, macrophages, monocytes, dendritic cells) depending on the conditions and can have either an inhibitory or stimulating orientation. Thus, endomorphins can be promising compounds that can effectively regulate both nociceptive signals and processes in the immune system.
Collapse
Affiliation(s)
- Sergey V Gein
- Institute of ecology and genetics of microorganisms - branch of the Perm Federal Research Center of the Ural Branch of the Russian Academy of Sciences; Perm State University
| | - Tatyana A Baeva
- Institute of ecology and genetics of microorganisms - branch of the Perm Federal Research Center of the Ural Branch of the Russian Academy of Sciences
| |
Collapse
|
7
|
Okano T, Sato K, Shirai R, Seki T, Shibata K, Yamashita T, Koide A, Tezuka H, Mori Y, Hirano T, Watanabe T. β-Endorphin Mediates the Development and Instability of Atherosclerotic Plaques. Int J Endocrinol 2020; 2020:4139093. [PMID: 32308678 PMCID: PMC7142353 DOI: 10.1155/2020/4139093] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 02/28/2020] [Indexed: 12/19/2022] Open
Abstract
β-Endorphin, an endogenous opioid peptide, and its μ-opioid receptor are expressed in brain, liver, and peripheral tissues. β-Endorphin induces endothelial dysfunction and is related to insulin resistance. We clarified the effects of β-endorphin on atherosclerosis. We assessed the effects of β-endorphin on the inflammatory response and monocyte adhesion in human umbilical vein endothelial cells (HUVECs), foam cell formation, and the inflammatory phenotype in THP-1 monocyte-derived macrophages, and migration and proliferation of human aortic smooth muscle cells (HASMCs) in vitro. We also assessed the effects of β-endorphin on aortic lesions in Apoe -/- mice in vivo. The μ-opioid receptor (OPRM1) was expressed in THP-1 monocytes, macrophages, HASMCs, HUVECs, and human aortic endothelial cells. β-Endorphin significantly increased THP-1 monocyte adhesion to HUVECs and induced upregulation of intercellular adhesion molecule-1, vascular cell adhesion molecule-1, and E-selectin via nuclear factor-κB (NF-κB) and p38 phosphorylation in HUVECs. β-Endorphin significantly increased HUVEC proliferation and enhanced oxidized low-density lipoprotein-induced foam cell formation in macrophages. β-Endorphin also significantly shifted the macrophage phenotype to proinflammatory M1 rather than anti-inflammatory M2 via NF-κB phosphorylation during monocyte-macrophage differentiation and increased migration and apoptosis in association with c-jun-N-terminal kinase, p38, and NF-κB phosphorylation in HASMCs. Chronic β-endorphin infusion into Apoe -/- mice significantly aggravated the development of aortic atherosclerotic lesions, with an increase in vascular inflammation and the intraplaque macrophage/smooth muscle cell ratio, an index of plaque instability. Our study provides the first evidence that β-endorphin contributes to the acceleration of the progression and instability of atheromatous plaques. Thus, μ-opioid receptor antagonists may be useful for the prevention and treatment of atherosclerosis.
Collapse
Affiliation(s)
- Taisuke Okano
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Kengo Sato
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
- Division of Laboratory and Transfusion Medicine, Hokkaido University Hospital, Kita-15 Nishi-7, Kita-ku, Sapporo, Hokkaido 060-8638, Japan
| | - Remina Shirai
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Tomomi Seki
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Koichiro Shibata
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Tomoyuki Yamashita
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Ayaka Koide
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Hitomi Tezuka
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Yusaku Mori
- Division of Diabetes, Metabolism, and Endocrinology, Department of Medicine, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8666, Japan
| | - Tsutomu Hirano
- Division of Diabetes, Metabolism, and Endocrinology, Department of Medicine, Showa University School of Medicine, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8666, Japan
| | - Takuya Watanabe
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
- Department of Internal Medicine, Ushioda General Hospital/Clinic, Yokohama, Japan
| |
Collapse
|
8
|
|
9
|
Böttcher B, Seeber B, Leyendecker G, Wildt L. Impact of the opioid system on the reproductive axis. Fertil Steril 2017; 108:207-213. [DOI: 10.1016/j.fertnstert.2017.06.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 06/06/2017] [Indexed: 12/15/2022]
|
10
|
Neuron-restrictive silencer factor-mediated downregulation of μ-opioid receptor contributes to the reduced morphine analgesia in bone cancer pain. Pain 2017; 158:879-890. [PMID: 28415063 PMCID: PMC5402709 DOI: 10.1097/j.pain.0000000000000848] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Neuron-restrictive silencer factor–induced downregulation of μ-opioid receptor is involved in the reduction of morphine analgesia in sarcoma-induced bone cancer pain. Bone cancer pain has been reported to have unique mechanisms and is resistant to morphine treatment. Recent studies have indicated that neuron-restrictive silencer factor (NRSF) plays a crucial role in modulating the expression of the μ-opioid receptor (MOR) gene. The present study elucidates the regulatory mechanisms of MOR and its ability to affect bone cancer pain. Using a sarcoma-inoculated murine model, pain behaviors that represent continuous or breakthrough pain were evaluated. Expression of NRSF in the dorsal root ganglion (DRG) and spinal dorsal horn was quantified at the transcriptional and translational levels, respectively. Additionally, chromatin immunoprecipitation assays were used to detect NRSF binding to the promoter of MOR. Furthermore, NRSF was genetically knocked out by antisense oligodeoxynucleotide, and the expression of MOR and the effect of morphine were subsequently analyzed. Our results indicated that in a sarcoma murine model, NRSF expression is upregulated in dorsal root ganglion neurons, and the expression of NRSF mRNA is significantly negatively correlated with MOR mRNA expression. Additionally, chromatin immunoprecipitation analysis revealed that NRSF binding to the neuron-restrictive silencer element within the promoter area of the MOR gene is promoted with a hypoacetylation state of histone H3 and H4. Furthermore, genetically knocking down NRSF with antisense oligodeoxynucleotide rescued the expression of MOR and potentiated the systemic morphine analgesia. The present results suggest that in sarcoma-induced bone cancer pain, NRSF-induced downregulation of MOR is involved in the reduction of morphine analgesia. Epigenetically, up-regulation of MOR could substantially improve the effect of system delivery of morphine.
Collapse
|
11
|
Zhang WP, Zong QF, Gao Q, Yu Y, Gu XY, Wang Y, Li ZH, Ge M. Effects of endomorphin-1 postconditioning on myocardial ischemia/reperfusion injury and myocardial cell apoptosis in a rat model. Mol Med Rep 2016; 14:3992-8. [DOI: 10.3892/mmr.2016.5695] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2015] [Accepted: 07/29/2016] [Indexed: 11/06/2022] Open
|
12
|
Opiate receptor blockade on human granulosa cells inhibits VEGF release. Reprod Biomed Online 2016; 32:316-22. [DOI: 10.1016/j.rbmo.2015.12.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Revised: 12/17/2015] [Accepted: 12/22/2015] [Indexed: 12/22/2022]
|
13
|
Affiliation(s)
- W Mahbuba
- Department of Cardiovascular Sciences, (Division of Anaesthesia, Critical Care and Pain Management), University of Leicester, Leicester Royal Infirmary, Leicester LE2 7LX, UK Department of Surgery, College of Medicine, University of Kufa, Kufa, Iraq
| | - D G Lambert
- Department of Cardiovascular Sciences, (Division of Anaesthesia, Critical Care and Pain Management), University of Leicester, Leicester Royal Infirmary, Leicester LE2 7LX, UK
| |
Collapse
|
14
|
Zhang YF, Xu QX, Liao LD, Xu XE, Wu JY, Wu ZY, Shen JH, Li EM, Xu LY. Association of mu-opioid receptor expression with lymph node metastasis in esophageal squamous cell carcinoma. Dis Esophagus 2015; 28:196-203. [PMID: 24428760 DOI: 10.1111/dote.12165] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The mu-opioid receptor (MOR), a membrane-bound G protein-coupled receptor, is the main target for opioids in the nervous system. MOR1 has been found in several types of cancer cells and reported to be involved in tumor progression and metastasis. However, the expression and clinical significance of MOR1 in esophageal squamous cell carcinoma (ESCC) remain unclear. In our study, the expression of MOR1 was confirmed in ESCC cell lines (KYSE180, KYSE150, and EC109) by Western blot. MOR1 was also detected on tissue microarrays of ESCC samples in 239 cases using immunohistochemical staining. We found that MOR1 was mainly located in the cytoplasm and occasionally occurred in the membrane or nucleus of ESCC cells. Moreover, results indicated that MOR1 expression in the cytoplasm was associated with lymph node metastasis (R = 0.164, P = 0.008, Kendall's tau-b-test). No more associations were found between MOR1 expression status and other clinical parameters. However, no statistical significant differences were found between MOR1 expression in the cytoplasm, nucleus/membrane, and the overall survival of ESCC patients (P = 0.848; P = 0.167; P = 0.428, respectively, log-rank test). Our results suggest that the cytoplasmic MOR1 may be a high-risk factor for lymph node metastasis of ESCC patients. We also hypothesize that MOR1 agonists used in ESCC patients should be prudent, and opioid receptor antagonists may be novel therapeutic drugs for ESCC patients.
Collapse
Affiliation(s)
- Y-F Zhang
- Institute of Oncologic Pathology, Shantou University Medical College, Shantou, China
| | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Meloxicam and buprenorphine treatment after ovarian transplantation does not affect estrous cyclicity and follicular integrity in aged CBA/J mice. PLoS One 2014; 9:e106013. [PMID: 25153315 PMCID: PMC4143324 DOI: 10.1371/journal.pone.0106013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Accepted: 07/28/2014] [Indexed: 11/29/2022] Open
Abstract
Angiogenesis, the formation of new blood vessels, is important for the survival of ovarian transplants and the restoration of ovarian functions. Without angiogenesis, transplanted ovarian tissue becomes more susceptible to tissue damage and necrosis. Administration of analgesics for pain management has been shown to decrease angiogenesis, which can influence transplant success especially in aged animals. Aging and the effects of hypoxia after transplantation decrease reproductive viability of the ovarian transplant; therefore, it is important to understand the additional effects of analgesics on aged animal models. The present study investigated the effects of two analgesics, buprenorphine, an opiate, and meloxicam, a non-steroidal anti-inflammatory drug (NSAID), on the reproductive indicators related to estrous cyclicity and follicular integrity after ovarian transplantation of young ovaries into aged CBA/J mice. These aged females did not show any different reproductive responses when treated with either buprenorphine or meloxicam. No significant differences were observed in estrous cycle length, the onset of estrous cycling, the regularity of estrous cycles, and the proportion of viable follicles and total number of follicles per ovarian sample across treatment groups.
Collapse
|
16
|
Bigliardi PL, Neumann C, Teo YL, Pant A, Bigliardi-Qi M. Activation of the δ-opioid receptor promotes cutaneous wound healing by affecting keratinocyte intercellular adhesion and migration. Br J Pharmacol 2014; 172:501-14. [PMID: 24628261 PMCID: PMC4292964 DOI: 10.1111/bph.12687] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Revised: 02/28/2014] [Accepted: 03/06/2014] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND AND PURPOSE In addition to its analgesic functions, the peripheral opioid receptor system affects skin homeostasis by influencing cell differentiation, migration and adhesion; also, wound healing is altered in δ-opioid receptor knockout mice (DOPr–/–). Hence, we investigated δ-opioid receptor effects on the expression of several proteins of the desmosomal junction complex and on the migratory behaviour of keratinocytes. EXPERIMENTAL APPROACH Expression levels of desmosomal cadherins in wild-type and DOPr–/– mice, and the morphology of intercellular adhesion in human keratinocytes were analysed by immunofluorescence. To investigate the δ-opioid receptor activation pathway, protein expression was studied using Western blot and its effect on cellular migration determined by in vitro live cell migration recordings from human keratinocytes. KEY RESULTS Expression of the desmosomal cadherins, desmogleins 1 and 4, was up-regulated in skin from DOPr–/– mice, and down-regulated in δ-opioid receptor-overexpressing human keratinocytes. The localization of desmoplakin expression was rearranged from linear arrays emanating from cell borders to puncta in cell periphery, resulting in less stable intercellular adhesion. Migration and wound recovery were enhanced in human keratinocyte monolayers overexpressing δ-opioid receptors in vitro. These δ-opioid receptor effects were antagonized by specific PKCα/β inhibition indicating they were mediated through the PKC signalling pathway. Finally, cells overexpressing δ-opioid receptors developed characteristically long but undirected protrusions containing filamentous actin and δ-opioid receptors, indicating an enhanced migratory phenotype. CONCLUSION AND IMPLICATIONS Opioid receptors affect intercellular adhesion and wound healing mechanisms, underlining the importance of a cutaneous neuroendocrine system in wound healing and skin homeostasis. LINKED ARTICLES This article is part of a themed section on Opioids: New Pathways to Functional Selectivity. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2015.172.issue-2
Collapse
Affiliation(s)
- P L Bigliardi
- Institute of Medical Biology, Agency for Science Technology & Research (A*STAR), Singapore; Division of Rheumatology, National University Hospital, University Medicine Cluster, Singapore
| | | | | | | | | |
Collapse
|
17
|
A new chiral 2-(ethylthio)-thiazolone analogue shows strong antitumor activities by inducing cancer cell apoptosis and inhibiting angiogenesis. Anticancer Drugs 2012; 23:914-22. [PMID: 22614105 DOI: 10.1097/cad.0b013e328354dc85] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Our initial study showed the potent cytotoxic effects of a series of new chiral 2-(ethylthio)-thiazolone analogues we synthesized. In the present study, we used computer prediction and found that nitro functionality and the modification of substituents R could further improve their activities in the presence of the nitro group. Compound 1s with nitro, naphthyl, ethyl groups, and a chiral center was predicted to be the most effective. We showed that compound 1s could inhibit the growth of five different cancer cell lines in a time-dependent and dose-dependent manner. 1s could induce Hela cell apoptosis by activating the mitochondria apoptotic pathway. In addition, 1s could inhibit the proliferation, migration, tuber formation, and adhesion of human umbilical vein endothelial cells, suggesting its antiangiogenesis effects. Furthermore, we confirmed the in-vivo antitumor effects of 1s on sarcoma S-180-bearing mice. Taken together, chiral 2-(ethylthio)-thiazolone analogue 1s is a promising compound for further anticancer drug development.
Collapse
|
18
|
Song H, Yin W, Zeng Q, Jia H, Lin L, Liu X, Mu L, Wang R. Hemokinins modulate endothelium function and promote angiogenesis through neurokinin-1 receptor. Int J Biochem Cell Biol 2012; 44:1410-21. [PMID: 22554585 DOI: 10.1016/j.biocel.2012.04.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2012] [Revised: 03/29/2012] [Accepted: 04/18/2012] [Indexed: 01/26/2023]
Abstract
Substance P as a member of tachykinin family plays an important role in angiogenesis. Hemokinins (HKs) have been identified as new members of substance P-like peptides of tachykinin family. However, the effects of HKs on endothelial cells and angiogenesis have not been studied. For the first time, here we demonstrated that r/mHK-1, hHK-1 and hHK(4-11) dose-dependently stimulated the proliferation, migration, adhesion and tube formation of freshly isolated human umbilical vein endothelial cells (HUVECs), and further exhibited in vivo angiogenic effects in chick embryo chorioallantoic membrane model. The angiogenic effects of HKs were inhibited by the selective antagonist of neurokinin-1 rather than neurokinin-2 receptor. Mechanistically, HKs activated ERK1/2 phosphorylation, stimulated nitric oxide production, and upregulated the expression of endothelial nitric oxide synthase (eNOS) and vascular endothelial growth factor (VEGF) in HUVECs. Taken together, our data suggest that HKs emerge as pivotal endogenous regulators of angiogenesis and represent potential targets for the intervention of angiogenesis in different pathological conditions given their specific peripheral distribution.
Collapse
Affiliation(s)
- Hongjin Song
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou, PR China.
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Lu D, Xia Y, Tong B, Zhang C, Pan R, Xu H, Yang X, Dai Y. In vitro anti-angiogenesis effects and active constituents of the saponin fraction from Gleditsia sinensis. Integr Cancer Ther 2012; 13:446-57. [PMID: 22505594 DOI: 10.1177/1534735412442377] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND The anomalous fruits of Gleditsia sinensis Lam. (Leguminosae), a crude drug in China, have long been used in traditional Chinese medicine for the treatment of various diseases. The saponin fraction isolated from the fruits (SFGS) is considered as the active component for the antitumor activity of this crude drug. OBJECTIVES The present study was performed to investigate the anti-angiogenesis activities and active constituents of SFGS. METHODS Human umbilical vein endothelial cells (HUVECs) were treated with SFGS in the presence or absence of basic fibroblast growth factor (bFGF) in vitro. The proliferation, migration, and tube formation were studied by MTT, Transwell, and 2D Matrigel assays, respectively. The cell cycle and apoptosis were analyzed by flow cytometry. Enzyme-linked immunosorbent assay for protein expression of vascular endothelial growth factor (VEGF) and western blot analysis for caspase-3, caspase-8, and caspase-9 as well as Fas were performed. In addition, the effects of 13 saponin compounds isolated from SFGS on the tube formation of HUVECs were screened, and the structure-activity relationships were discussed. RESULTS SFGS, at concentrations (1, 3, and 10 µg/mL) without significant cytotoxicity on endothelial cells, significantly inhibited the proliferation, migration, and tube formation of HUVECs induced by bFGF (10 ng/mL). It moderately arrested the cell cycle to G1 phase but greatly induced cell apoptosis and increased the expressions of caspases-3, caspase-8, and Fas but not caspase-9 in HUVECs. Moreover, SFGS did not affect the bFGF-induced autosecretion of VEGF from endothelial cells. Among the 13 saponin compounds tested, gleditsiosides B, I, J, O, and Q showed inhibition of the tube formation at a concentration of 3 µM, and only gleditsioside B exerted significant inhibition at 1 µM. CONCLUSION SFGS is substantially able to prevent angiogenesis by interfering with multiple steps. The findings provide a new explanation for the antitumor effects of G sinensis fruits. Gleditsiosides B, I, J, O, and Q are probably the main active constituents of SFGS.
Collapse
Affiliation(s)
- Dan Lu
- China Pharmaceutical University, Nanjing, China
| | - YuFeng Xia
- China Pharmaceutical University, Nanjing, China
| | - Bei Tong
- China Pharmaceutical University, Nanjing, China
| | | | - Rong Pan
- China Pharmaceutical University, Nanjing, China
| | - Huan Xu
- China Pharmaceutical University, Nanjing, China
| | - Xue Yang
- China Pharmaceutical University, Nanjing, China
| | - Yue Dai
- China Pharmaceutical University, Nanjing, China
| |
Collapse
|
20
|
Influence of morphine on pericyte-endothelial interaction: implications for antiangiogenic therapy. JOURNAL OF ONCOLOGY 2012; 2012:458385. [PMID: 22315595 PMCID: PMC3270445 DOI: 10.1155/2012/458385] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2011] [Accepted: 10/07/2011] [Indexed: 12/12/2022]
Abstract
Morphine stimulates tumor angiogenesis and cancer progression in mice. We examined if morphine influences endothelial-pericyte interaction via platelet-derived growth factor-BB (PDGF-BB) and PDGF receptor-β (PDGFR-β). Clinically relevant doses of morphine stimulated PDGF-BB secretion from human umbilical vein endothelial cells and activated PDGFR-β and mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) phosphorylation in human pericytes. These in vitro effects of morphine were translated into promotion of tumor angiogenesis in a transgenic mice model of breast cancer when treated with clinically used dose of morphine. Increased vessel-associated immunoreactivity of desmin and PDGFR-β was observed on pericytes in tumors of morphine-treated mice. These data suggest that morphine potentiates endothelial-pericyte interaction via PDGF-BB/PDGFR-β signaling and promotes tumor angiogenesis, pericyte recruitment, and coverage of tumor vessels. We speculate that morphine may impair the effectiveness of antiangiogenic therapy by influencing vascular pericyte coverage.
Collapse
|
21
|
Abstract
This paper is the thirty-third consecutive installment of the annual review of research concerning the endogenous opioid system. It summarizes papers published during 2010 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides, opioid receptors, opioid agonists and opioid antagonists. The particular topics that continue to be covered include the molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors related to behavior (Section 2), and the roles of these opioid peptides and receptors in pain and analgesia (Section 3); stress and social status (Section 4); tolerance and dependence (Section 5); learning and memory (Section 6); eating and drinking (Section 7); alcohol and drugs of abuse (Section 8); sexual activity and hormones, pregnancy, development and endocrinology (Section 9); mental illness and mood (Section 10); seizures and neurologic disorders (Section 11); electrical-related activity and neurophysiology (Section 12); general activity and locomotion (Section 13); gastrointestinal, renal and hepatic functions (Section 14); cardiovascular responses (Section 15); respiration (Section 16); and immunological responses (Section 17).
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Psychology and Neuropsychology Doctoral Sub-Program, Queens College, City University of New York, Flushing, NY 11367, USA.
| |
Collapse
|
22
|
Abstract
Morphine is an analgesic widely used to alleviate cancer pain. In addition, the perioperative management of pain in cancer surgery patients most often includes opioids. However, there are reports that these drugs may alter cancer recurrence or metastasis. Several mechanisms have been proposed, such as the modulation of the immune response or cellular pathways that control the survival and migratory behavior of cancer cells. The published literature, however, presents some discrepancies, with reports suggesting that opioids may either promote or prevent the spread of cancer. It is of great importance to determine whether opioids, in particular the most widely used, morphine, may increase the risk of metastasis when used in cancer surgery. This review examines the available data on the effects of morphine which influence cancer metastasis or recurrence, including immunomodulation, tumor cell aggressiveness, and angiogenesis, with special emphasis on recently published clinical and laboratory based studies. We further discuss the parameters that may explain the difference between reports on the effects of morphine on cancer.
Collapse
|
23
|
Liu J, Wei S, Tian L, Yan L, Guo Q, Ma X. Effects of endomorphins on human umbilical vein endothelial cells under high glucose. Peptides 2011; 32:86-92. [PMID: 20970471 DOI: 10.1016/j.peptides.2010.09.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2010] [Revised: 09/27/2010] [Accepted: 09/27/2010] [Indexed: 12/19/2022]
Abstract
The endomorphin-1 (EM1) and endomorphin-2 (EM2) are endogenous opioid peptides, which modulate extensive bioactivities such as pain, cardiovascular responses, immunological responses and so on. The present study was undertaken to investigate the effects of EM1/EM2 on the primary cultured human umbilical vein endothelial cells (HUVECs) damaged by high glucose. PI AnnexinV-FITC detection was performed to evaluate the apoptosis rate. Levels of nitric oxide (NO) and nitric oxide synthase (NOS) activity were measured by the Griess reaction and the conversion of 3H-arginine to 3H-citrulline, respectively. Endothelin-1 (ET-1) was evaluated by the enzyme-linked immunosorbent assay (ELISA). Cell proliferation was determined by the MTT viability assay. mRNA expression of endothelial nitric oxide synthase (eNOS) and ET-1 were measured by real-time PCR. Our data showed that EM1/EM2 inhibited cell apoptosis. The high glucose induced increase in expression of NO, NOS and ET-1 were significantly attenuated by pretreatment with EM1/EM2 in a dose dependent manner. In addition, EM1/EM2 suppressed the mRNA eNOS and mRNA ET-1 expression in HUVECs under high glucose conditions. Naloxone, the nonselective opioid receptor antagonist, did not influence the mRNA eNOS expression when it was administrated on its own; but it could significantly antagonize the effects induced by EM1/EM2. Furthermore, in all assay systems, EM1 was more potent than EM2. The results suggest that EM1/EM2 have a beneficial effect in protecting against the endothelial dysfunction by high glucose in vitro, and these effects were mediated by the opioid receptors in HUVECs.
Collapse
Affiliation(s)
- Jing Liu
- Department of Endocrinology, the People's Hospital of Gansu Province, 204 Donggang West Road, Lanzhou 730000, PR China.
| | | | | | | | | | | |
Collapse
|