1
|
Lee HJ, Kim SH, Kim YH, Kim SH, Oh GS, Bae JE, Kim JB, Park NY, Park K, Yeom E, Jeong K, Kim P, Jo DS, Cho DH. Nalfurafine Hydrochloride, a κ-Opioid Receptor Agonist, Induces Melanophagy via PKA Inhibition in B16F1 Cells. Cells 2022; 12:cells12010146. [PMID: 36611940 PMCID: PMC9818167 DOI: 10.3390/cells12010146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/19/2022] [Accepted: 12/28/2022] [Indexed: 01/01/2023] Open
Abstract
Selective autophagy controls cellular homeostasis by degrading unnecessary or damaged cellular components. Melanosomes are specialized organelles that regulate the biogenesis, storage, and transport of melanin in melanocytes. However, the mechanisms underlying melanosomal autophagy, known as the melanophagy pathway, are poorly understood. To better understand the mechanism of melanophagy, we screened an endocrine-hormone chemical library and identified nalfurafine hydrochlorides, a κ-opioid receptor agonist, as a potent inducer of melanophagy. Treatment with nalfurafine hydrochloride increased autophagy and reduced melanin content in alpha-melanocyte-stimulating hormone (α-MSH)-treated cells. Furthermore, inhibition of autophagy blocked melanosomal degradation and reversed the nalfurafine hydrochloride-induced decrease in melanin content in α-MSH-treated cells. Consistently, treatment with other κ-opioid receptor agonists, such as MCOPPB or mianserin, inhibited excessive melanin production but induced autophagy in B16F1 cells. Furthermore, nalfurafine hydrochloride inhibited protein kinase A (PKA) activation, which was notably restored by forskolin, a PKA activator. Additionally, forskolin treatment further suppressed melanosomal degradation as well as the anti-pigmentation activity of nalfurafine hydrochloride in α-MSH-treated cells. Collectively, our data suggest that stimulation of κ-opioid receptors induces melanophagy by inhibiting PKA activation in α-MSH-treated B16F1 cells.
Collapse
Affiliation(s)
- Ha Jung Lee
- BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Seong Hyun Kim
- BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Yong Hwan Kim
- BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, Kyungpook National University, Daegu 41566, Republic of Korea
| | - So Hyun Kim
- BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Gyeong Seok Oh
- BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Ji-Eun Bae
- Brain Science and Engineering Institute, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Joon Bum Kim
- BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Na Yeon Park
- BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Kyuhee Park
- Bio-center, Gyeonggido Business & Science Accelerator, Gyeonggido, Suwon 16229, Republic of Korea
| | - Eunbyul Yeom
- BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Kwiwan Jeong
- Bio-center, Gyeonggido Business & Science Accelerator, Gyeonggido, Suwon 16229, Republic of Korea
| | - Pansoo Kim
- Bio-center, Gyeonggido Business & Science Accelerator, Gyeonggido, Suwon 16229, Republic of Korea
| | - Doo Sin Jo
- BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, Kyungpook National University, Daegu 41566, Republic of Korea
- Correspondence: (D.S.J.); (D.-H.C.); Tel.: +82-53-950-5382 (D.S.J. & D.-H.C.)
| | - Dong-Hyung Cho
- BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, Kyungpook National University, Daegu 41566, Republic of Korea
- OGASIS Corp. 260, Changyong-daero, Yongtong-gu, Suwon 08826, Republic of Korea
- Correspondence: (D.S.J.); (D.-H.C.); Tel.: +82-53-950-5382 (D.S.J. & D.-H.C.)
| |
Collapse
|
2
|
Dalefield ML, Scouller B, Bibi R, Kivell BM. The Kappa Opioid Receptor: A Promising Therapeutic Target for Multiple Pathologies. Front Pharmacol 2022; 13:837671. [PMID: 35795569 PMCID: PMC9251383 DOI: 10.3389/fphar.2022.837671] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 05/20/2022] [Indexed: 11/13/2022] Open
Abstract
Kappa-opioid receptors (KOR) are widely expressed throughout the central nervous system, where they modulate a range of physiological processes depending on their location, including stress, mood, reward, pain, inflammation, and remyelination. However, clinical use of KOR agonists is limited by adverse effects such as dysphoria, aversion, and sedation. Within the drug-development field KOR agonists have been extensively investigated for the treatment of many centrally mediated nociceptive disorders including pruritis and pain. KOR agonists are potential alternatives to mu-opioid receptor (MOR) agonists for the treatment of pain due to their anti-nociceptive effects, lack of abuse potential, and reduced respiratory depressive effects, however, dysphoric side-effects have limited their widespread clinical use. Other diseases for which KOR agonists hold promising therapeutic potential include pruritis, multiple sclerosis, Alzheimer's disease, inflammatory diseases, gastrointestinal diseases, cancer, and ischemia. This review highlights recent drug-development efforts targeting KOR, including the development of G-protein-biased ligands, mixed opioid agonists, and peripherally restricted ligands to reduce side-effects. We also highlight the current KOR agonists that are in preclinical development or undergoing clinical trials.
Collapse
Affiliation(s)
| | | | | | - Bronwyn M. Kivell
- Centre for Biodiscovery, School of Biological Sciences, Victoria University of Wellington, Wellington, New Zealand
| |
Collapse
|
3
|
Endogenous Opioids and Their Role in Stem Cell Biology and Tissue Rescue. Int J Mol Sci 2022; 23:ijms23073819. [PMID: 35409178 PMCID: PMC8998234 DOI: 10.3390/ijms23073819] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 03/28/2022] [Accepted: 03/29/2022] [Indexed: 01/25/2023] Open
Abstract
Opioids are considered the oldest drugs known by humans and have been used for sedation and pain relief for several centuries. Nowadays, endogenous opioid peptides are divided into four families: enkephalins, dynorphins, endorphins, and nociceptin/orphanin FQ. They exert their action through the opioid receptors (ORs), transmembrane proteins belonging to the super-family of G-protein-coupled receptors, and are expressed throughout the body; the receptors are the δ opioid receptor (DOR), μ opioid receptor (MOR), κ opioid receptor (KOR), and nociceptin/orphanin FQ receptor (NOP). Endogenous opioids are mainly studied in the central nervous system (CNS), but their role has been investigated in other organs, both in physiological and in pathological conditions. Here, we revise their role in stem cell (SC) biology, since these cells are a subject of great scientific interest due to their peculiar features and their involvement in cell-based therapies in regenerative medicine. In particular, we focus on endogenous opioids’ ability to modulate SC proliferation, stress response (to oxidative stress, starvation, or damage following ischemia–reperfusion), and differentiation towards different lineages, such as neurogenesis, vasculogenesis, and cardiogenesis.
Collapse
|
4
|
Huang HM, He XH, Huang XY, Wang GY, Xia QX, Du ZP, Zhang YF. Down-regulation of kappa opioid receptor promotes ESCC proliferation, invasion and metastasis via the PDK1-AKT signaling pathway. Cell Commun Signal 2022; 20:35. [PMID: 35305679 PMCID: PMC8934502 DOI: 10.1186/s12964-022-00833-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 01/29/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND As a class of the opioid receptors, the kappa opioid receptor (KOR) has been verified to be a potential biomarker and therapeutic target for human malignant tumors. However, a thorough understanding of whether KOR affects progression of esophageal squamous cell carcinoma (ESCC) is still lacking. This study focused on exploring the effect of knocking down KOR in ESCC and its underlying mechanism. METHODS Bioinformatics analysis was used to compare the different expression level of OPRK1 (KOR gene) in tumor and adjacent normal tissues, and predict the relationship between KOR expression and overall survival. RNA-sequence analysis was performed to detect the altered functions and mechanisms after down regulating KOR. The in vitro and in vivo assays were used to detect the effects of down-regulated KOR on cell proliferation, migration and invasion. Substrate gel zymography and 3D cell culture assays were used to find the effect of KOR knockdown on the degradation of extracellular matrix (ECM), and immunefluorescence was performed to detect the altered cytoskeleton. Western blotting and immunohistochemistry were used to explore the underlying mechanism pathway. RESULTS Bioinformatics analysis revealed that the expression of OPRK1 was lower in tumor tissue than that in adjacent normal tissues, and lowered expression of KOR was associated with poorer overall survival. The in vitro assays demonstrated that down-regulation of KOR enhanced ESCC proliferation, metastasis and invasion. Western blotting revealed that down-regulation of KOR could activate PDK1-AKT signaling pathway, which actively regulated the cancer progression. Down-regulation of KOR enhanced the formation of invadopodia, secretion of matrix metalloproteinase-2 (MMP2) and rearrangement of cytoskeleton, which were positively related with the invasion of ESCC. KOR knockdown enhanced the tumor invasion and elevated the AKT phosphorylation in nude mice. The AKT kinase inhibition could reverse the effect of down-regulation of KOR. CONCLUSION KOR might act as a tumor suppressor in ESCC and down-regulation of KOR could enhance the ESCC tumor phenotype. Video Abstract.
Collapse
Affiliation(s)
- Han-Ming Huang
- Department of Anesthesiology, Second Affiliated Hospital of Shantou University Medical College, Shantou, 515041, People's Republic of China
| | - Xin-Hua He
- Department of Physiology, Shantou University Medical College, Shantou, 515041, People's Republic of China
| | - Xiao-Yu Huang
- Department of Anesthesiology, Second Affiliated Hospital of Shantou University Medical College, Shantou, 515041, People's Republic of China
| | - Guo-Yun Wang
- Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, 518067, People's Republic of China
| | - Qiao-Xi Xia
- Central Laboratory, Shantou Central Hospital, Shantou, 515041, People's Republic of China
| | - Ze-Peng Du
- Central Laboratory, Shantou Central Hospital, Shantou, 515041, People's Republic of China.
- Department of Pathology, Shantou Central Hospital, Shantou, 515041, People's Republic of China
| | - Yong-Fa Zhang
- Department of Anesthesiology, Second Affiliated Hospital of Shantou University Medical College, Shantou, 515041, People's Republic of China
| |
Collapse
|
5
|
Gopalakrishnan L, Chatterjee O, Ravishankar N, Suresh S, Raju R, Mahadevan A, Prasad TSK. Opioid receptors signaling network. J Cell Commun Signal 2021; 16:475-483. [PMID: 34724150 DOI: 10.1007/s12079-021-00653-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 10/12/2021] [Indexed: 10/19/2022] Open
Abstract
Opioid receptors belong to the class A G-protein-coupled receptors and are activated by alkaloid opiates such as morphine, and endogenous ligands such as endorphins and enkephalins. Opioid receptors are widely distributed in the human body and are involved in numerous physiological processes through three major classical opioid receptor subtypes; the mu, delta and kappa along with a lesser characterized subtype, opioid receptor-like (ORL1). Opioids are the most potent analgesics and have been extensively used as a therapeutic drug for the treatment of pain and related disorders. Chronic administration of clinically used opioids is associated with adverse effects such as drug tolerance, addiction and constipation. Several investigations attempted to identify the molecular signaling networks associated with endogenous as well as synthetic opiates, however, there is a paucity of a cumulative depiction of these signaling events. Here, we report a systemic collection of downstream molecules pertaining to four subtypes of opioid receptors (MOR, KOR, DOR and ORL1) in the form of a signaling pathway map. We manually curated reactions induced by the activation of opioid receptors from the literature into five categories- molecular association, activation/inhibition, catalysis, transport, and gene regulation. This led to a dataset of 180 molecules, which is collectively represented in the opioid receptor signaling network following NetPath criteria. We believe that the public availability of an opioid receptor signaling pathway map can accelerate biomedical research in this area because of its high therapeutic significance. The opioid receptors signaling pathway map is uploaded to a freely available web resource, WikiPathways enabling ease of access ( https://www.wikipathways.org/index.php/Pathway:WP5093 ).
Collapse
Affiliation(s)
- Lathika Gopalakrishnan
- Institute of Bioinformatics, International Tech Park, Bangalore, 560 066, India.,Manipal Academy of Higher Education (MAHE), Manipal, 576 104, India.,Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed To Be University), Mangalore, 575 018, India
| | - Oishi Chatterjee
- Institute of Bioinformatics, International Tech Park, Bangalore, 560 066, India.,Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed To Be University), Mangalore, 575 018, India.,Amrita School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam, 690 525, India
| | - Namitha Ravishankar
- Institute of Bioinformatics, International Tech Park, Bangalore, 560 066, India
| | - Sneha Suresh
- Institute of Bioinformatics, International Tech Park, Bangalore, 560 066, India
| | - Rajesh Raju
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed To Be University), Mangalore, 575 018, India.
| | - Anita Mahadevan
- Department of Neuropathology, National Institute of Mental Health and Neurosciences, Bangalore, 560 029, India.,Human Brain Tissue Repository, National Institute of Mental Health and Neurosciences, Neurobiology Research Centre, Bangalore, 560 029, India
| | - T S Keshava Prasad
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed To Be University), Mangalore, 575 018, India.
| |
Collapse
|
6
|
Cissom C, J Paris J, Shariat-Madar Z. Dynorphins in Development and Disease: Implications for Cardiovascular Disease. Curr Mol Med 2021; 20:259-274. [PMID: 31746302 DOI: 10.2174/1566524019666191028122559] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Revised: 10/18/2019] [Accepted: 10/18/2019] [Indexed: 12/12/2022]
Abstract
It is well-established that cardiovascular disease continues to represent a growing health problem and significant effort has been made to elucidate the underlying mechanisms. In this review, we report on past and recent high impact publications in the field of intracrine network signaling, focusing specifically on opioids and their interrelation with key modulators of the cardiovascular system and the onset of related disease. We present an overview of studies outlining the scope of cardiovascular and cerebrovascular processes that are affected by opioids, including heart function, ischemia, reperfusion, and blood flow. Specific emphasis is placed on the importance of dynorphin molecules in cerebrovascular and cardiovascular regulation. Evidence suggests that excessive or insufficient dynorphin could make an important contribution to cardiovascular physiology, yet numerous paradoxical observations frequently impede a clear understanding of the role of dynorphin. Thus, we argue that dynorphin-mediated signaling events for which an immediate regulatory effect is disputed should not be dismissed as unimportant, as they may play a role in cross-talk with other signaling networks. Finally, we consider the most recent evidence on the role of dynorphin during cardiovascular-related inflammation and on the potential value of endogenous and exogenous inhibitors of kappa-opioid receptor, a major dynorphin A receptor, to limit or prevent cardiovascular disease and its related sequelae.
Collapse
Affiliation(s)
- Cody Cissom
- William Carey College of Osteopathic Mississippi University, Medical School, Hattiesburg, Mississippi, United States
| | - Jason J Paris
- Department of Biomolecular Sciences, Division of Pharmacology, University of Mississippi, United States.,The National Center for Natural Products Research, Research Institute of Pharmaceutical Sciences, University of Mississippi, United States
| | - Zia Shariat-Madar
- Department of Biomolecular Sciences, Division of Pharmacology, University of Mississippi, United States.,The National Center for Natural Products Research, Research Institute of Pharmaceutical Sciences, University of Mississippi, United States.,Light Microscopy Core, University of Mississippi, University, Mississippi, United States
| |
Collapse
|
7
|
Shokirova H, Inomata T, Saitoh T, Zhu J, Fujio K, Okumura Y, Yanagawa A, Fujimoto K, Sung J, Eguchi A, Miura M, Nagino K, Hirosawa K, Kuwahara M, Akasaki Y, Nagase H, Murakami A. Topical administration of the kappa opioid receptor agonist nalfurafine suppresses corneal neovascularization and inflammation. Sci Rep 2021; 11:8647. [PMID: 33883646 PMCID: PMC8060258 DOI: 10.1038/s41598-021-88118-6] [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: 12/05/2020] [Accepted: 04/07/2021] [Indexed: 12/11/2022] Open
Abstract
Corneal neovascularization (CNV) causes higher-order aberrations, corneal edema, ocular inflammation, and corneal transplant rejection, thereby decreasing visual acuity. In this study, we investigated the effects of topical administration of the kappa opioid receptor agonist nalfurafine (TRK-820) on CNV. To induce CNV, intrastromal corneal sutures were placed on the corneal stroma of BALB/c mice for 2 weeks. Nalfurafine (0.1 µg/2 μL/eye) was topically administered to the cornea once or twice daily after CNV induction. The CNV score, immune cell infiltration, and mRNA levels of angiogenic and pro-inflammatory factors in neovascularized corneas were evaluated using slit-lamp microscopy, immunohistochemistry, flow cytometry, and polymerase chain reaction. The mRNA expression of the kappa opioid receptor gene Oprk1 was significantly upregulated following CNV induction. Topical administration of nalfurafine twice daily significantly suppressed CNV and lymphangiogenesis, as well as reduced the mRNA levels of angiogenic and pro-inflammatory factors in the neovascularized corneas. Moreover, nalfurafine administration twice daily reduced the numbers of infiltrating leukocytes, neutrophils, macrophages, and interferon-γ-producing CD4+ T cells in the neovascularized corneas. In this study, we demonstrated that topical administration of nalfurafine suppressed local CNV in a mouse model along with the activation of KOR, suggesting that nalfurafine may prevent and control CNV in humans.
Collapse
Affiliation(s)
- Hurramhon Shokirova
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, 3-1-3 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Takenori Inomata
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, 3-1-3 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan. .,Department of Ophthalmology, Juntendo University Faculty of Medicine, Tokyo, Japan. .,Department of Strategic Operating Room Management and Improvement, Juntendo University Graduate School of Medicine, Tokyo, Japan. .,Department of Hospital Administration, Juntendo University Graduate School of Medicine, Tokyo, Japan. .,Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan.
| | - Tsuyoshi Saitoh
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, 1-1-1 Tennodai, Ibaraki, Japan
| | - Jun Zhu
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, 3-1-3 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.,Department of Ophthalmology, Subei People's Hospital Affiliated to Yangzhou University, Yangzhou, Jiangsu Province, China
| | - Kenta Fujio
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, 3-1-3 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.,Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yuichi Okumura
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, 3-1-3 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.,Department of Strategic Operating Room Management and Improvement, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Ai Yanagawa
- Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Keiichi Fujimoto
- Department of Ophthalmology, Juntendo University Faculty of Medicine, Tokyo, Japan.,Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Jaemyoung Sung
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, 3-1-3 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Atsuko Eguchi
- Department of Hospital Administration, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Maria Miura
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, 3-1-3 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.,Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Ken Nagino
- Department of Hospital Administration, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Kunihiko Hirosawa
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, 3-1-3 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.,Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Mizu Kuwahara
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, 3-1-3 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.,Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yasutsugu Akasaki
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, 3-1-3 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.,Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hiroshi Nagase
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, 1-1-1 Tennodai, Ibaraki, Japan
| | - Akira Murakami
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, 3-1-3 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.,Department of Ophthalmology, Juntendo University Faculty of Medicine, Tokyo, Japan.,Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
8
|
Shi Y, Luo J, Tian J, Zou Q, Wang X. The kappa opioid receptor may be a potential tumor suppressor by regulating angiogenesis in breast cancer. Med Hypotheses 2021; 150:110568. [PMID: 33780776 DOI: 10.1016/j.mehy.2021.110568] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 02/14/2021] [Accepted: 03/09/2021] [Indexed: 01/15/2023]
Abstract
Our hypothesis proposes that activating κ-opioid receptors (KORs) may inhibit the progression of breast cancer and improve patient prognosis. Consequently, KORs may become a promising therapeutic target for breast cancer. Activating KORs induces not only analgesic efficacy comparable to μ-opioid receptors but also shows a promising antitumor effect and with fewer opioid-induced adverse effects. Based on present studies and our bioinformatics analysis of KORs, we propose that KORs can function as a tumor suppressor by inhibiting angiogenesis in human breast cancer; therefore, analgesics that mainly activate KORs would be more suitable for breast cancer patients.
Collapse
Affiliation(s)
- Yumiao Shi
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai 200127, China
| | - Jiamei Luo
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai 200127, China
| | - Jie Tian
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai 200127, China
| | - Qiaoqun Zou
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai 200127, China.
| | - Xiaoqiang Wang
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai 200127, China.
| |
Collapse
|
9
|
Canaider S, Facchin F, Tassinari R, Cavallini C, Olivi E, Taglioli V, Zannini C, Bianconi E, Maioli M, Ventura C. Intracrine Endorphinergic Systems in Modulation of Myocardial Differentiation. Int J Mol Sci 2019; 20:ijms20205175. [PMID: 31635381 PMCID: PMC6829321 DOI: 10.3390/ijms20205175] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 10/11/2019] [Accepted: 10/16/2019] [Indexed: 12/12/2022] Open
Abstract
A wide variety of peptides not only interact with the cell surface, but govern complex signaling from inside the cell. This has been referred to as an "intracrine" action, and the orchestrating molecules as "intracrines". Here, we review the intracrine action of dynorphin B, a bioactive end-product of the prodynorphin gene, on nuclear opioid receptors and nuclear protein kinase C signaling to stimulate the transcription of a gene program of cardiogenesis. The ability of intracrine dynorphin B to prime the transcription of its own coding gene in isolated nuclei is discussed as a feed-forward loop of gene expression amplification and synchronization. We describe the role of hyaluronan mixed esters of butyric and retinoic acids as synthetic intracrines, controlling prodynorphin gene expression, cardiogenesis, and cardiac repair. We also discuss the increase in prodynorphin gene transcription and intracellular dynorphin B afforded by electromagnetic fields in stem cells, as a mechanism of cardiogenic signaling and enhancement in the yield of stem cell-derived cardiomyocytes. We underline the possibility of using the diffusive features of physical energies to modulate intracrinergic systems without the needs of viral vector-mediated gene transfer technologies, and prompt the exploration of this hypothesis in the near future.
Collapse
Affiliation(s)
- Silvia Canaider
- National Laboratory of Molecular Biology and Stem Cell Bioengineering - Eldor Lab, National Institute of Biostructures and Biosystems (NIBB), at the Innovation Accelerator, CNR, Via Piero Gobetti 101, 40129 Bologna, Italy.
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy.
| | - Federica Facchin
- National Laboratory of Molecular Biology and Stem Cell Bioengineering - Eldor Lab, National Institute of Biostructures and Biosystems (NIBB), at the Innovation Accelerator, CNR, Via Piero Gobetti 101, 40129 Bologna, Italy.
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy.
| | - Riccardo Tassinari
- National Laboratory of Molecular Biology and Stem Cell Bioengineering - Eldor Lab, National Institute of Biostructures and Biosystems (NIBB), at the Innovation Accelerator, CNR, Via Piero Gobetti 101, 40129 Bologna, Italy.
| | - Claudia Cavallini
- National Laboratory of Molecular Biology and Stem Cell Bioengineering - Eldor Lab, National Institute of Biostructures and Biosystems (NIBB), at the Innovation Accelerator, CNR, Via Piero Gobetti 101, 40129 Bologna, Italy.
| | - Elena Olivi
- National Laboratory of Molecular Biology and Stem Cell Bioengineering - Eldor Lab, National Institute of Biostructures and Biosystems (NIBB), at the Innovation Accelerator, CNR, Via Piero Gobetti 101, 40129 Bologna, Italy.
| | - Valentina Taglioli
- National Laboratory of Molecular Biology and Stem Cell Bioengineering - Eldor Lab, National Institute of Biostructures and Biosystems (NIBB), at the Innovation Accelerator, CNR, Via Piero Gobetti 101, 40129 Bologna, Italy.
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy.
| | - Chiara Zannini
- National Laboratory of Molecular Biology and Stem Cell Bioengineering - Eldor Lab, National Institute of Biostructures and Biosystems (NIBB), at the Innovation Accelerator, CNR, Via Piero Gobetti 101, 40129 Bologna, Italy.
| | - Eva Bianconi
- National Laboratory of Molecular Biology and Stem Cell Bioengineering - Eldor Lab, National Institute of Biostructures and Biosystems (NIBB), at the Innovation Accelerator, CNR, Via Piero Gobetti 101, 40129 Bologna, Italy.
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy.
| | - Margherita Maioli
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy.
| | - Carlo Ventura
- National Laboratory of Molecular Biology and Stem Cell Bioengineering - Eldor Lab, National Institute of Biostructures and Biosystems (NIBB), at the Innovation Accelerator, CNR, Via Piero Gobetti 101, 40129 Bologna, Italy.
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy.
| |
Collapse
|
10
|
Reddy LVK, Sen D. Regulation of Cardiomyocyte Differentiation, Angiogenesis, and Inflammation by the Delta Opioid Signaling in Human Mesenchymal Stem Cells. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2019. [DOI: 10.1007/s40883-019-00100-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
11
|
Šínová R, Kudová J, Nešporová K, Karel S, Šuláková R, Velebný V, Kubala L. Opioid receptors and opioid peptides in the cardiomyogenesis of mouse embryonic stem cells. J Cell Physiol 2018; 234:13209-13219. [DOI: 10.1002/jcp.27992] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 11/21/2018] [Indexed: 12/15/2022]
Affiliation(s)
- Romana Šínová
- Department of Free Radical Pathophysiology Institute of Biophysics, Academy of Sciences of the Czech Republic Brno Czech Republic
- Institute of Experimental Biology, Faculty of Science, Masaryk University Brno Czech Republic
- Contipro a. s. Dolni Dobrouc Czech Republic
| | - Jana Kudová
- Department of Free Radical Pathophysiology Institute of Biophysics, Academy of Sciences of the Czech Republic Brno Czech Republic
| | | | | | | | | | - Lukáš Kubala
- Department of Free Radical Pathophysiology Institute of Biophysics, Academy of Sciences of the Czech Republic Brno Czech Republic
- Institute of Experimental Biology, Faculty of Science, Masaryk University Brno Czech Republic
- International Clinical Research Center, St. Anne' University Hospital Brno Czech Republic
| |
Collapse
|
12
|
Byrnes EM, Vassoler FM. Modeling prenatal opioid exposure in animals: Current findings and future directions. Front Neuroendocrinol 2018; 51:1-13. [PMID: 28965857 PMCID: PMC5649358 DOI: 10.1016/j.yfrne.2017.09.001] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 09/26/2017] [Accepted: 09/27/2017] [Indexed: 01/03/2023]
Abstract
The past decade has seen a drastic rise in the number of infants exposed to opioids in utero. It is unclear what lasting effect this exposure may have on these children. Animal models of prenatal opioid exposure may provide insight into potential areas of vulnerability. The present review summarizes the findings across animal models of prenatal opioid exposure, including exposure to morphine, methadone, buprenorphine, and oxycodone. Details regarding the drug, doses, and duration of treatment, as well as key findings, are summarized in tables with associated references. Finally, significant gaps in the current preclinical literature and future directions are discussed.
Collapse
Affiliation(s)
- Elizabeth M Byrnes
- Department of Biomedical Sciences, Cummings School of Veterinary Medicine, Tufts University, North Grafton, MA 01536, United States.
| | - Fair M Vassoler
- Department of Biomedical Sciences, Cummings School of Veterinary Medicine, Tufts University, North Grafton, MA 01536, United States
| |
Collapse
|
13
|
Yang BZ, Han S, Kranzler HR, Palmer AA, Gelernter J. Sex-specific linkage scans in opioid dependence. Am J Med Genet B Neuropsychiatr Genet 2017; 174:261-268. [PMID: 27762075 PMCID: PMC5695218 DOI: 10.1002/ajmg.b.32507] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 10/03/2016] [Indexed: 02/01/2023]
Abstract
Sex influences risk for opioid dependence (OD). We hypothesized that sex might interact with genetic loci that influence the risk for OD. Therefore we performed an analysis to identify sex-specific genomic susceptibility regions for OD using linkage. Over 6,000 single nucleotide polymorphism (SNP) markers were genotyped for 1,758 African- and European-American (AA and EA) individuals from 739 families, ascertained via affected sib-pairs with OD and/or cocaine dependence. Autosomewide non-parametric linkage scans, stratified by sex and population, were performed. We identified one significant linkage region, segregating with OD in EA men, at 71.1 cM on chromosome 4 (LOD = 3.29; point-wise P = 0.00005; empirical autosome-wide P = 0.042), which significantly differed from the linkage signal at the same location in EA women (empirical P = 0.002). Three suggestive linkage signals were identified at 181.3 cM on chromosome 7 (LOD = 2.18), 104 cM on chromosome 11 (LOD = 1.85), and 60.9 cM on chromosome 16 (LOD = 1.93) in EA women. In AA men, four suggestive linkage signals were detected at 201.1 cM on chromosome 3 (LOD = 2.32), 152.9 cM on chromosome 6 (LOD = 1.86), 16.8 cM on chromosome 7 (LOD = 1.95), and 36.1 cM on chromosome 17 (LOD = 1.99). The significant region, mapping to 4q12-4q13.1, harbors several OD candidate genes with interconnected functionality, including VEGFR, CLOCK, PDCL2, NMU, NRSF, and IGFBP7. In conclusion, these results provide an evidence for the existence of sex-specific and population-specific differences in OD. Furthermore, these results provide positional information that will facilitate the use of targeted next-generation sequencing to search for genes that contribute to sex-specific differences in OD. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Bao-Zhu Yang
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT
| | - Shizhong Han
- Department of Psychiatry, University of Iowa, Iowa City, IA
| | - Henry R. Kranzler
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine and VISN 4 MIRECC, Crescenz VAMC, Philadelphia, PA
| | - Abraham A. Palmer
- Department of Psychiatry, University of California San Diego, La Jolla, CA
| | - Joel Gelernter
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT,VA CT Healthcare Center, West Haven, CT
| |
Collapse
|
14
|
Wu L, Zhang S, Shkhyan R, Lee S, Gullo F, Eliasberg CD, Petrigliano FA, Ba K, Wang J, Lin Y, Evseenko D. Kappa opioid receptor signaling protects cartilage tissue against posttraumatic degeneration. JCI Insight 2017; 2:e88553. [PMID: 28097228 DOI: 10.1172/jci.insight.88553] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Osteoarthritis is the most common form of arthritis, and pain relief with opioid-like drugs is a commonly used therapeutic for osteoarthritic patients. Recent studies published by our group showed that the kappa opioid receptor (KOR) is highly expressed during human development in joint-forming cells. However, the precise role of this receptor in the skeletal system remains elusive. The main aim of the current study was to investigate the role of KOR signaling in synovial and cartilaginous tissues in pathological conditions. Our data demonstrate that KOR null mice exhibit accelerated cartilage degeneration after injury when compared with WT mice. Activation of KOR signaling increased the expression of anabolic enzymes and inhibited cartilage catabolism and degeneration in response to proinflammatory cytokines such as TNF-α. In addition, selective KOR agonists increased joint lubrication via the activation of cAMP/CREB signaling in chondrocytes and synovial cells. Taken together, these results demonstrate direct effects of KOR agonists on cartilage and synovial cells and reveals a protective effect of KOR signaling against cartilage degeneration after injury. In addition to pain control, local administration of dynorphin or other KOR agonist represents an attractive therapeutic approach in patients with early stages of osteoarthritis.
Collapse
Affiliation(s)
- Ling Wu
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Shu Zhang
- State Key Laboratory for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Ruzanna Shkhyan
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Siyoung Lee
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Francesca Gullo
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Claire D Eliasberg
- Department of Orthopaedic Surgery, David Geffen School of Medicine, UCLA, Los Angeles, California, USA
| | - Frank A Petrigliano
- Department of Orthopaedic Surgery, David Geffen School of Medicine, UCLA, Los Angeles, California, USA
| | - Kai Ba
- State Key Laboratory for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Jing Wang
- Department of Stomatology, Tenth People's Hospital of Tongji University, Shanghai, China
| | - Yunfeng Lin
- State Key Laboratory for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Denis Evseenko
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
15
|
Opioid κ Receptors as a Molecular Target for the Creation of a New Generation of Analgesic Drugs. Pharm Chem J 2016. [DOI: 10.1007/s11094-016-1388-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
16
|
Aguirre JA, Lucchinetti E, Clanachan AS, Plane F, Zaugg M. Unraveling Interactions Between Anesthetics and the Endothelium. Anesth Analg 2016; 122:330-48. [DOI: 10.1213/ane.0000000000001053] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
17
|
Banerjee J, Papu John AMS, Schuller HM. Regulation of nonsmall-cell lung cancer stem cell like cells by neurotransmitters and opioid peptides. Int J Cancer 2015; 137:2815-24. [PMID: 26088878 DOI: 10.1002/ijc.29646] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Revised: 05/07/2015] [Accepted: 05/29/2015] [Indexed: 12/21/2022]
Abstract
Nonsmall-cell lung cancer (NSCLC) is the leading type of lung cancer and has a poor prognosis. We have shown that chronic stress promoted NSCLC xenografts in mice via stress neurotransmitter-activated cAMP signaling downstream of beta-adrenergic receptors and incidental beta-blocker therapy was reported to improve clinical outcomes in NSCLC patients. These findings suggest that psychological stress promotes NSCLC whereas pharmacologically or psychologically induced decreases in cAMP may inhibit NSCLC. Cancer stem cells are thought to drive the development, progression and resistance to therapy of NSCLC. However, their potential regulation by stress neurotransmitters has not been investigated. In the current study, epinephrine increased the number of cancer stem cell like cells (CSCs) from three NSCLC cell lines in spheroid formation assays while enhancing intracellular cAMP and the stem cell markers sonic hedgehog (SHH), aldehyde dehydrogenase-1 (ALDH-1) and Gli1, effects reversed by GABA or dynorphin B via Gαi -mediated inhibition of cAMP formation. The growth of NSCLC xenografts in a mouse model of stress reduction was significantly reduced as compared with mice maintained under standard conditions. Stress reduction reduced serum levels of corticosterone, norepinephrine and epinephrine while the inhibitory neurotransmitter γ-aminobutyric acid (GABA) and opioid peptides increased. Stress reduction significantly reduced cAMP, VEGF, p-ERK, p-AKT, p-CREB, p-SRc, SHH, ALDH-1 and Gli1 in xenograft tissues whereas cleaved caspase-3 and p53 were induced. We conclude that stress neurotransmitters activate CSCs in NSCLC via multiple cAMP-mediated pathways and that pharmacologically or psychologically induced decreases in cAMP signaling may improve clinical outcomes in NSCLC patients.
Collapse
Affiliation(s)
- Jheelam Banerjee
- Experimental Oncology Laboratory, Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN
| | - Arokya M S Papu John
- Experimental Oncology Laboratory, Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN
| | - Hildegard M Schuller
- Experimental Oncology Laboratory, Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN
| |
Collapse
|
18
|
Egleton RD, Abbruscato T. Drug abuse and the neurovascular unit. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2014; 71:451-80. [PMID: 25307226 DOI: 10.1016/bs.apha.2014.06.019] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Drug abuse continues to create a major international epidemic affecting society. A great majority of past drug abuse research has focused mostly on the mechanisms of addiction and the specific effects of substance use disorders on brain circuits and pathways that modulate reward, motivation, craving, and decision making. Few studies have focused on the neurobiology of acute and chronic substance abuse as it relates to the neurovascular unit (brain endothelial cell, neuron, astrocyte, microglia, and pericyte). Increasing research indicates that all cellular components of the neurovascular unit play a pivotal role in both the process of addiction and how drug abuse affects the brain response to diseases. This review will focus on the specific effects of opioids, amphetamines, alcohol, and nicotine on the neurovascular unit and its role in addiction and adaption to brain diseases. Elucidation of the role of the neurovascular unit on the neurobiology associated with drug addiction will help to facilitate the development of better therapeutic approaches for drug-dependent individuals.
Collapse
Affiliation(s)
- Richard D Egleton
- Department of Pharmacology, Physiology and Toxicology, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia, USA.
| | - Thomas Abbruscato
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas, USA.
| |
Collapse
|
19
|
Hulse RP, Beazley-Long N, Hua J, Kennedy H, Prager J, Bevan H, Qiu Y, Fernandes ES, Gammons MV, Ballmer-Hofer K, Gittenberger de Groot AC, Churchill AJ, Harper SJ, Brain SD, Bates DO, Donaldson LF. Regulation of alternative VEGF-A mRNA splicing is a therapeutic target for analgesia. Neurobiol Dis 2014; 71:245-59. [PMID: 25151644 PMCID: PMC4194316 DOI: 10.1016/j.nbd.2014.08.012] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Revised: 07/29/2014] [Accepted: 08/06/2014] [Indexed: 12/02/2022] Open
Abstract
Vascular endothelial growth factor-A (VEGF-A) is best known as a key regulator of the formation of new blood vessels. Neutralization of VEGF-A with anti-VEGF therapy e.g. bevacizumab, can be painful, and this is hypothesized to result from a loss of VEGF-A-mediated neuroprotection. The multiple vegf-a gene products consist of two alternatively spliced families, typified by VEGF-A165a and VEGF-A165b (both contain 165 amino acids), both of which are neuroprotective. Under pathological conditions, such as in inflammation and cancer, the pro-angiogenic VEGF-A165a is upregulated and predominates over the VEGF-A165b isoform. We show here that in rats and mice VEGF-A165a and VEGF-A165b have opposing effects on pain, and that blocking the proximal splicing event – leading to the preferential expression of VEGF-A165b over VEGF165a – prevents pain in vivo. VEGF-A165a sensitizes peripheral nociceptive neurons through actions on VEGFR2 and a TRPV1-dependent mechanism, thus enhancing nociceptive signaling. VEGF-A165b blocks the effect of VEGF-A165a. After nerve injury, the endogenous balance of VEGF-A isoforms switches to greater expression of VEGF-Axxxa compared to VEGF-Axxxb, through an SRPK1-dependent pre-mRNA splicing mechanism. Pharmacological inhibition of SRPK1 after traumatic nerve injury selectively reduced VEGF-Axxxa expression and reversed associated neuropathic pain. Exogenous VEGF-A165b also ameliorated neuropathic pain. We conclude that the relative levels of alternatively spliced VEGF-A isoforms are critical for pain modulation under both normal conditions and in sensory neuropathy. Altering VEGF-Axxxa/VEGF-Axxxb balance by targeting alternative RNA splicing may be a new analgesic strategy. The different vegf-a splice variants, VEGF-A165a and VEGF-A165b have pro- and anti-nociceptive actions respectively. Pro-nociceptive actions of VEGF-A165a are dependent on TRPV1. Alternative pre-mRNA splicing underpins peripheral sensitization by VEGF-A isoforms in normal and neuropathic animals.
Collapse
Affiliation(s)
- R P Hulse
- Physiology and Pharmacology, University of Bristol, Bristol BS8 1TD, UK; Cancer Biology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Queen's Medical Centre, Nottingham NG2 7UH, UK
| | - N Beazley-Long
- Physiology and Pharmacology, University of Bristol, Bristol BS8 1TD, UK; School of Life Sciences, The Medical School, University of Nottingham, Queen's Medical Centre, Nottingham NG2 7UH, UK
| | - J Hua
- Physiology and Pharmacology, University of Bristol, Bristol BS8 1TD, UK
| | - H Kennedy
- Physiology and Pharmacology, University of Bristol, Bristol BS8 1TD, UK
| | - J Prager
- Physiology and Pharmacology, University of Bristol, Bristol BS8 1TD, UK
| | - H Bevan
- Physiology and Pharmacology, University of Bristol, Bristol BS8 1TD, UK
| | - Y Qiu
- Physiology and Pharmacology, University of Bristol, Bristol BS8 1TD, UK
| | | | - M V Gammons
- Physiology and Pharmacology, University of Bristol, Bristol BS8 1TD, UK
| | | | | | - A J Churchill
- Clinical Sciences, University of Bristol, Bristol BS1 2LX, UK
| | - S J Harper
- Physiology and Pharmacology, University of Bristol, Bristol BS8 1TD, UK
| | - S D Brain
- King's College London, London SE1 9NH, UK
| | - D O Bates
- Cancer Biology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Queen's Medical Centre, Nottingham NG2 7UH, UK.
| | - L F Donaldson
- Physiology and Pharmacology, University of Bristol, Bristol BS8 1TD, UK; School of Life Sciences, The Medical School, University of Nottingham, Queen's Medical Centre, Nottingham NG2 7UH, UK.
| |
Collapse
|
20
|
Yamamizu K, Hamada Y, Narita M. κ Opioid receptor ligands regulate angiogenesis in development and in tumours. Br J Pharmacol 2014; 172:268-76. [PMID: 24417697 DOI: 10.1111/bph.12573] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Revised: 12/09/2013] [Accepted: 01/04/2014] [Indexed: 12/20/2022] Open
Abstract
UNLABELLED Opioid systems mainly regulate physiological functions such as pain, emotional tone and reward circuitry in neural tissues (brain and spinal cord). These systems are also found in extraneural tissues (ganglia, gut, spleen, stomach, lung, pancreas, liver, heart, blood and blood vessels), and recent studies have elucidated their roles in various organs. The current review focuses on the roles of opioid systems in blood vessels, especially angiogenesis, during development and tumour malignancy. The balance between endogenous activators and inhibitors of angiogenesis delicately maintains a normally quiescent vasculature to sustain homeostasis. Disturbance of this balance causes pathogenic angiogenesis and, especially in tumours, several activators such as VEGF are highly expressed in the tumour microenvironment and strongly induce tumour angiogenesis, the so-called angiogenic switch. Recently, we demonstrated that κ opioid receptor agonists function as anti-angiogenic factors, which impede the angiogenic switch, in vascular development and tumour angiogenesis by inhibiting the expression of receptors for VEGF. In clinical medicine, angiogenesis inhibitors that target VEGF signalling such as bevacizumab are used as anti-cancer drugs. Although therapies that inhibit tumour angiogenesis have been highly successful for tumour therapy, most patients eventually develop resistance to this anti-angiogenic therapy. Thus, we must identify novel targets for anti-angiogenic agents to sustain inhibition of angiogenesis for tumour therapy. The regulation of responses to κ opioid receptor ligands could be useful for controlling vascular formation under physiological conditions and in cancers, and thus could offer therapeutic benefits beyond the relief of pain. 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)
- Kohei Yamamizu
- Laboratory of Genetics, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | | | | |
Collapse
|
21
|
Ma J, Wan J, Meng J, Banerjee S, Ramakrishnan S, Roy S. Methamphetamine induces autophagy as a pro-survival response against apoptotic endothelial cell death through the Kappa opioid receptor. Cell Death Dis 2014; 5:e1099. [PMID: 24603327 PMCID: PMC3973232 DOI: 10.1038/cddis.2014.64] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Revised: 01/27/2014] [Accepted: 01/28/2014] [Indexed: 11/09/2022]
Abstract
Methamphetamine (METH) is a psychostimulant with high abuse potential and severe neurotoxicity. Recent studies in animal models have indicated that METH can impair the blood-brain barrier (BBB), suggesting that some of the neurotoxic effects resulting from METH abuse could be due to barrier disruption. We report here that while chronic exposure to METH disrupts barrier function of primary human brain microvascular endothelial cells (HBMECs) and human umbilical vein endothelial cells (HUVECs), an early pro-survival response is observed following acute exposure by induction of autophagic mechanisms. Acute METH exposure induces an early increase in Beclin1 and LC3 recruitment. This is mediated through inactivation of the protein kinase B (Akt)/mammalian target of rapamycin (mTOR)/p70S6K pathway, and upregulation of the ERK1/2. Blockade of Kappa opioid receptor (KOR), and treatment with autophagic inhibitors accelerated METH-induced apoptosis, suggesting that the early autophagic response is a survival mechanism for endothelial cells and is mediated through the kappa opioid receptor. Our studies indicate that kappa opioid receptor can be therapeutically exploited for attenuating METH-induced BBB dysfunction.
Collapse
Affiliation(s)
- J Ma
- Departments of Surgery, University of Minnesota, Minneapolis, MN, USA
| | - J Wan
- Departments of Surgery, University of Minnesota, Minneapolis, MN, USA
| | - J Meng
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, USA
| | - S Banerjee
- Departments of Surgery, University of Minnesota, Minneapolis, MN, USA
| | - S Ramakrishnan
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, USA
| | - S Roy
- Departments of Surgery, University of Minnesota, Minneapolis, MN, USA
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
22
|
Single-step generation of gene knockout-rescue system in pluripotent stem cells by promoter insertion with CRISPR/Cas9. Biochem Biophys Res Commun 2014; 444:158-63. [DOI: 10.1016/j.bbrc.2014.01.037] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Accepted: 01/09/2014] [Indexed: 11/21/2022]
|
23
|
Yamamizu K, Furuta S, Hamada Y, Yamashita A, Kuzumaki N, Narita M, Doi K, Katayama S, Nagase H, Yamashita JK, Narita M. к Opioids inhibit tumor angiogenesis by suppressing VEGF signaling. Sci Rep 2013; 3:3213. [PMID: 24225480 PMCID: PMC3827603 DOI: 10.1038/srep03213] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Accepted: 10/28/2013] [Indexed: 01/09/2023] Open
Abstract
Opioids are effective analgesics for the management of moderate to severe cancer pain. Here we show that κ opioid receptor (KOR) agonists act as anti-angiogenic factors in tumors. Treatment with KOR agonists, U50,488H and TRK820, significantly inhibited human umbilical vein endothelial cell (HUVEC) migration and tube formation by suppressing VEGFR2 expression. In contrast, treatment with a μ opioid receptor agonist, DAMGO, or a δ opioid receptor agonist, SNC80, did not prevent angiogenesis in HUVECs. Lewis lung carcinoma (LLC) or B16 melanoma grafted in KOR knockout mice showed increased proliferation and remarkably enhanced tumor angiogenesis compared with those in wild type mice. On the other hand, repeated intraperitoneal injection of TRK820 (0.1-10 μg/kg, b.i.d.) significantly inhibited tumor growth by suppressing tumor angiogenesis. These findings indicate that KOR agonists play an important role in tumor angiogenesis and this knowledge could lead to a novel strategy for cancer therapy.
Collapse
Affiliation(s)
- Kohei Yamamizu
- 1] Laboratory of Stem Cell Differentiation, Stem Cell Research Center, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan [2] Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan [3] Laboratory of Genetics, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA [4]
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Nivet AL, Vigneault C, Blondin P, Sirard MA. Changes in granulosa cells' gene expression associated with increased oocyte competence in bovine. Reproduction 2013; 145:555-65. [PMID: 23564726 DOI: 10.1530/rep-13-0032] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
One of the challenges in mammalian reproduction is to understand the basic physiology of oocyte quality. It is believed that the follicle status is linked to developmental competence of the enclosed oocyte. To explore the link between follicles and competence in cows, previous research at our laboratory has developed an ovarian stimulation protocol that increases and then decreases oocyte quality according to the timing of oocyte recovery post-FSH withdrawal (coasting). Using this protocol, we have obtained the granulosa cells associated with oocytes of different qualities at selected times of coasting. Transcriptome analysis was done with Embryogene microarray slides and validation was performed by real-time PCR. Results show that the major changes in gene expression occurred from 20 to 44 h of coasting, when oocyte quality increases. Secondly, among upregulated genes (20-44 h), 25% were extracellular molecules, highlighting potential granulosa signaling cascades. Principal component analysis identified two patterns: one resembling the competence profile and another associated with follicle growth and atresia. Additionally, three major functional changes were identified: (i) the end of follicle growth (BMPR1B, IGF2, and RELN), involving interactions with the extracellular matrix (TFPI2); angiogenesis (NRP1), including early hypoxia, and potentially oxidative stress (GFPT2, TF, and VNN1) and (ii) apoptosis (KCNJ8) followed by iii) inflammation (ANKRD1). This unique window of analysis indicates a progressive hypoxia during coasting mixed with an increase in apoptosis and inflammation. Potential signaling pathways leading to competence have been identified and will require downstream testing. This preliminary analysis supports the potential role of the follicular differentiation in oocyte quality both during competence increase and decrease phases.
Collapse
Affiliation(s)
- Anne-Laure Nivet
- Département des sciences animales, Pavillon INAF, Faculté des sciences de l'agriculture et de l'alimentation, Centre de recherche en biologie de la reproduction, Université Laval, Quebec, Quebec, Canada G1V 0A6
| | | | | | | |
Collapse
|
25
|
Abstract
This paper is the thirty-fourth consecutive installment of the annual review of research concerning the endogenous opioid system. It summarizes papers published during 2011 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, United States.
| |
Collapse
|
26
|
López-Bellido R, Barreto-Valer K, Sánchez-Simón FM, Rodríguez RE. Cocaine modulates the expression of opioid receptors and miR-let-7d in zebrafish embryos. PLoS One 2012; 7:e50885. [PMID: 23226419 PMCID: PMC3511421 DOI: 10.1371/journal.pone.0050885] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2012] [Accepted: 10/25/2012] [Indexed: 11/19/2022] Open
Abstract
Prenatal exposure to cocaine, in mammals, has been shown to interfere with the expression of opioid receptors, which can have repercussions in its activity. Likewise, microRNAs, such as let-7, have been shown to regulate the expression of opioid receptors and hence their functions in mammals and in vitro experiments. In light of this, using the zebrafish embryos as a model our aim here was to evaluate the actions of cocaine in the expression of opioid receptors and let-7d miRNA during embryogenesis. In order to determine the effects produced by cocaine on the opioid receptors (zfmor, zfdor1 and zfdor2) and let-7d miRNA (dre-let-7d) and its precursors (dre-let-7d-1 and dre-let-7d-2), embryos were exposed to 1.5 µM cocaine hydrochloride (HCl). Our results revealed that cocaine upregulated dre-let-7d and its precursors, and also increased the expression of zfmor, zfdor1 and zfdor2 during early developmental stages and decreased them in late embryonic stages. The changes observed in the expression of opioid receptors might occur through dre-let-7d, since DNA sequences and the morpholinos of opioid receptors microinjections altered the expression of dre-let-7d and its precursors. Likewise, opioid receptors and dre-let-7d showed similar distributions in the central nervous system (CNS) and at the periphery, pointing to a possible interrelationship between them.In conclusion, the silencing and overexpression of opioid receptors altered the expression of dre-let-7d, which points to the notion that cocaine via dre-let-7 can modulate the expression of opioid receptors. Our study provides new insights into the actions of cocaine during zebrafish embryogenesis, indicating a role of miRNAs, let-7d, in development and its relationship with gene expression of opioid receptors, related to pain and addiction process.
Collapse
Affiliation(s)
- Roger López-Bellido
- Department of Biochemistry and Molecular Biology, Institute of Neuroscience of Castilla y León, University of Salamanca, Salamanca, Spain
| | - Katherine Barreto-Valer
- Department of Biochemistry and Molecular Biology, Institute of Neuroscience of Castilla y León, University of Salamanca, Salamanca, Spain
| | - Fátima Macho Sánchez-Simón
- Department of Biochemistry and Molecular Biology, Institute of Neuroscience of Castilla y León, University of Salamanca, Salamanca, Spain
| | - Raquel E. Rodríguez
- Department of Biochemistry and Molecular Biology, Institute of Neuroscience of Castilla y León, University of Salamanca, Salamanca, Spain
- * E-mail:
| |
Collapse
|
27
|
ROCK suppression promotes differentiation and expansion of endothelial cells from embryonic stem cell-derived Flk1(+) mesodermal precursor cells. Blood 2012; 120:2733-44. [PMID: 22896004 DOI: 10.1182/blood-2012-04-421610] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Successful differentiation and expansion of endothelial cells (ECs) from embryonic stem cell (ESC)-derived Flk1(+) mesodermal precursor cells (MPCs) requires supplementation of vascular endothelial growth factor-A (VEGF-A). While analyzing VEGF-A/VEGFR2 downstream signaling pathway that underlies the VEGF-A-induced differentiation and expansion of ECs, we fortuitously found that Rho-associated protein kinase (ROCK) inhibitor Y27632 profoundly promoted the differentiation and expansion of ECs from Flk1(+) MPCs while reducing the differentiation and expansion of mural cells. The ROCK suppression-induced expansion of ECs appears to have resulted from promotion of proliferation of ECs via activation of PI3-kinase-Akt signaling. The ECs obtained by the combination of ROCK suppression and VEGF-A supplementation faithfully expressed most pan-EC surface makers, and phenotypic analyses revealed that they were differentiated toward arterial EC. Further incubation of the ICAM2(+) ECs with Y27632 and VEGF-A for 2 days promoted expansion of ECs by 6.5-fold compared with those incubated with only VEGF-A. Importantly, the ROCK suppression-induced ECs displayed neovasculogenic abilities in vitro and in vivo. Thus, supplementation of ROCK inhibitor Y27632 along with VEGF-A in 2D Matrigel culture system provides a simple, efficient, and versatile method for obtaining ample amount of ESC-derived ECs at high purity suitable for use in therapeutic neovascularization.
Collapse
|
28
|
Yamamizu K, Matsunaga T, Katayama S, Kataoka H, Takayama N, Eto K, Nishikawa SI, Yamashita JK. PKA/CREB signaling triggers initiation of endothelial and hematopoietic cell differentiation via Etv2 induction. Stem Cells 2012; 30:687-96. [PMID: 22267325 DOI: 10.1002/stem.1041] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Ets family protein Etv2 (also called ER71 or Etsrp) is a key factor for initiation of vascular and blood development from mesodermal cells. However, regulatory mechanisms and inducing signals for Etv2 expression have been largely unknown. Previously, we revealed that cyclic adenosine monophosphate (cAMP)/protein kinase A (PKA) signaling enhanced differentiation of vascular progenitors into endothelial cells (ECs) and hematopoietic cells (HPCs) using an embryonic stem cell (ESC) differentiation system. Here, we show that PKA activation in an earlier differentiation stage can trigger EC/HPC differentiation through Etv2 induction. We found Etv2 was markedly upregulated by PKA activation preceding EC and HPC differentiation. We identified two cAMP response element (CRE) sequences in the Etv2 promoter and 5'-untranslated region and confirmed that CRE-binding protein (CREB) directly binds to the CRE sites and activates Etv2 transcription. Expression of a dominant negative form of CREB completely inhibited PKA-elicited Etv2 expression and induction of EC/HPCs from ESCs. Furthermore, blockade of PKA significantly inhibited Etv2 expression in ex vivo whole-embryo culture using Etv2-Venus knockin mice. These data indicated that PKA/CREB pathway is a critical regulator for the initiation of EC/HPC differentiation via Etv2 transcription. This early-stage molecular linkage between a triggering signal and transcriptional cascades for differentiation would provide novel insights in vascular and blood development and cell fate determination.
Collapse
Affiliation(s)
- Kohei Yamamizu
- Laboratory of Stem Cell Differentiation, Stem Cell Research Center, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan; Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Thomas RS, Black MB, Li L, Healy E, Chu TM, Bao W, Andersen ME, Wolfinger RD. A comprehensive statistical analysis of predicting in vivo hazard using high-throughput in vitro screening. Toxicol Sci 2012; 128:398-417. [PMID: 22543276 DOI: 10.1093/toxsci/kfs159] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Over the past 5 years, increased attention has been focused on using high-throughput in vitro screening for identifying chemical hazards and prioritizing chemicals for additional in vivo testing. The U.S. Environmental Protection Agency's ToxCast program has generated a significant amount of high-throughput screening data allowing a broad-based assessment of the utility of these assays for predicting in vivo responses. In this study, a comprehensive cross-validation model comparison was performed to evaluate the predictive performance of the more than 600 in vitro assays from the ToxCast phase I screening effort across 60 in vivo endpoints using 84 different statistical classification methods. The predictive performance of the in vitro assays was compared and combined with that from chemical structure descriptors. With the exception of chronic in vivo cholinesterase inhibition, the overall predictive power of both the in vitro assays and the chemical descriptors was relatively low. The predictive power of the in vitro assays was not significantly different from that of the chemical descriptors and aggregating the assays based on genes reduced predictive performance. Prefiltering the in vitro assay data outside the cross-validation loop, as done in some previous studies, significantly biased estimates of model performance. The results suggest that the current ToxCast phase I assays and chemicals have limited applicability for predicting in vivo chemical hazards using standard statistical classification methods. However, if viewed as a survey of potential molecular initiating events and interpreted as risk factors for toxicity, the assays may still be useful for chemical prioritization.
Collapse
Affiliation(s)
- Russell S Thomas
- The Hamner Institutes for Health Sciences Research Triangle Park, North Carolina 27709, USA.
| | | | | | | | | | | | | | | |
Collapse
|