1
|
Allen BG, Merlen C, Branco AF, Pétrin D, Hébert TE. Understanding the impact of nuclear-localized GPCRs on cellular signalling. Cell Signal 2024; 123:111358. [PMID: 39181220 DOI: 10.1016/j.cellsig.2024.111358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 08/14/2024] [Accepted: 08/20/2024] [Indexed: 08/27/2024]
Abstract
G protein-coupled receptors (GPCRs) have historically been associated with signalling events driven from the plasma membrane. More recently, signalling from endosomes has been recognized as a feature of internalizing receptors. However, there was little consideration given to the notion that GPCRs can be targeted to distinct subcellular locations that did not involve an initial trafficking to the cell surface. Here, we focus on the evidence for and the potential impact of GPCR signalling specifically initiated from the nuclear membrane. We also discuss the possibilities for selectively targeting this and other internal pools of receptors as novel venues for drug discovery.
Collapse
Affiliation(s)
- Bruce G Allen
- Montreal Heart Institute, Montréal, Québec H1T 1C8, Canada; Departments of Biochemistry and Molecular Medicine, Medicine, Pharmacology and Physiology, Université de Montréal, Montréal, Québec H3T 1J4, Canada
| | | | - Ana F Branco
- Montreal Heart Institute, Montréal, Québec H1T 1C8, Canada
| | - Darlaine Pétrin
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Terence E Hébert
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada.
| |
Collapse
|
2
|
Kang W, Siewe AD, Oluigbo CC, Arijesudade MO, Brailoiu E, Undieh AS. Dopamine internalization via Uptake 2 and stimulation of intracellular D 5-receptor-dependent calcium mobilization and CDP-diacylglycerol signaling. Front Pharmacol 2024; 15:1422998. [PMID: 39525629 PMCID: PMC11543475 DOI: 10.3389/fphar.2024.1422998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024] Open
Abstract
Dopamine stimulates CDP-diacylglycerol biosynthesis through D1-like receptors, particularly the D5 subtype most of which is intracellularly localized. CDP-diacylglycerol regulates phosphatidylinositol-4,5-bisphosphate-dependent signaling cascades by serving as obligatory substrate for phosphatidylinositol biosynthesis. Here, we used acute and organotypic brain tissues and cultured cells to explore the mechanism by which extracellular dopamine acts to modulate intracellular CDP-diacylglycerol. Dopamine stimulated CDP-diacylglycerol in organotypic and neural cells lacking the presynaptic dopamine transporter, and this action was selectively mimicked by D1-like receptor agonists SKF38393 and SKF83959. Dopaminergic CDP-diacylglycerol stimulation was blocked by decynium-22 which blocks Uptake2-like transporters and by anti-microtubule disrupters of cytoskeletal transport, suggesting transmembrane uptake and guided transport of the ligands to intracellular sites of CDP-diacylglycerol regulation. Fluorescent or radiolabeled dopamine was saturably transported into primary neurons or B35 neuroblastoma cells expressing the plasmamembrane monoamine transporter, PMAT. Microinjection of 10-nM final concentration of dopamine into human D5-receptor-transfected U2-OS cells rapidly and transiently increased cytosolic calcium concentrations by 316%, whereas non-D5-receptor-expressing U2-OS cells showed no response. Given that U2-OS cells natively express PMAT, bath application of 10 μM dopamine slowly increased cytosolic calcium in D5-expressing cells. These observations indicate that dopamine is actively transported by a PMAT-implicated Uptake2-like mechanism into postsynaptic-type dopaminoceptive cells where the monoamine stimulates its intracellular D5-type receptors to mobilize cytosolic calcium and promote CDP-diacylglycerol biosynthesis. This is probably the first demonstration of functional intracellular dopamine receptor coupling in neural tissue, thus challenging the conventional paradigm that postsynaptic dopamine uptake serves merely as a mechanism for deactivating spent or excessive synaptic transmitter.
Collapse
Affiliation(s)
- Wenfei Kang
- Department of Biomedical Sciences, School of Medicine, City University of New York, New York, NY, United States
| | - Arlette Deukam Siewe
- Department of Biomedical Sciences, School of Medicine, City University of New York, New York, NY, United States
| | - Chizurum C. Oluigbo
- Department of Biomedical Sciences, School of Medicine, City University of New York, New York, NY, United States
- Neuroscience Collaborative, The Graduate Center, City University of New York, New York, NY, United States
| | - Mercy O. Arijesudade
- Department of Biomedical Sciences, School of Medicine, City University of New York, New York, NY, United States
| | - Eugen Brailoiu
- Department of Pharmacology, School of Medicine, Temple University, Philadelphia, PA, United States
| | - Ashiwel S. Undieh
- Department of Biomedical Sciences, School of Medicine, City University of New York, New York, NY, United States
- Neuroscience Collaborative, The Graduate Center, City University of New York, New York, NY, United States
| |
Collapse
|
3
|
Wang CH, Tsuji T, Wu LH, Yang CY, Huang TL, Sato M, Shamsi F, Tseng YH. Endothelin 3/EDNRB signaling induces thermogenic differentiation of white adipose tissue. Nat Commun 2024; 15:7215. [PMID: 39174539 PMCID: PMC11341701 DOI: 10.1038/s41467-024-51579-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 08/12/2024] [Indexed: 08/24/2024] Open
Abstract
Thermogenic adipose tissue, consisting of brown and beige fat, regulates nutrient utilization and energy metabolism. Human brown fat is relatively scarce and decreases with obesity and aging. Hence, inducing thermogenic differentiation of white fat offers an attractive way to enhance whole-body metabolic capacity. Here, we show the role of endothelin 3 (EDN3) and endothelin receptor type B (EDNRB) in promoting the browning of white adipose tissue (WAT). EDNRB overexpression stimulates thermogenic differentiation of human white preadipocytes through cAMP-EPAC1-ERK activation. In mice, cold induces the expression of EDN3 and EDNRB in WAT. Deletion of EDNRB in adipose progenitor cells impairs cold-induced beige adipocyte formation in WAT, leading to excessive weight gain, glucose intolerance, and insulin resistance upon high-fat feeding. Injection of EDN3 into WAT promotes browning and improved whole-body glucose metabolism. The findings shed light on the mechanism of WAT browning and offer potential therapeutics for obesity and metabolic disorders.
Collapse
Affiliation(s)
- Chih-Hao Wang
- Graduate Institute of Cell Biology, China Medical University, Taichung City, Taiwan.
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung City, Taiwan.
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA.
| | - Tadataka Tsuji
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Li-Hong Wu
- Graduate Institute of Cell Biology, China Medical University, Taichung City, Taiwan
| | - Cheng-Ying Yang
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung City, Taiwan
| | - Tian Lian Huang
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Mari Sato
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Farnaz Shamsi
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, NY, USA
| | - Yu-Hua Tseng
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA.
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
4
|
Shi J, Jiao T, Guo Q, Weng W, Ma L, Zhang Q, Wang L, Zhang J, Chen C, Huang Y, Wang M, Pan R, Tang Y, Hu W, Meng T, Liu SH, Guo J, Kong Y, Meng X. A Cell Surface-Binding Antibody Atlas Nominates a MUC18-Directed Antibody-Drug Conjugate for Targeting Melanoma. Cancer Res 2023; 83:3783-3795. [PMID: 37668527 PMCID: PMC10646479 DOI: 10.1158/0008-5472.can-23-1356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 07/03/2023] [Accepted: 08/29/2023] [Indexed: 09/06/2023]
Abstract
Recent advances in targeted therapy and immunotherapy have substantially improved the treatment of melanoma. However, therapeutic strategies are still needed for unresponsive or treatment-relapsed patients with melanoma. To discover antibody-drug conjugate (ADC)-tractable cell surface targets for melanoma, we developed an atlas of melanoma cell surface-binding antibodies (pAb) using a proteome-scale antibody array platform. Target identification of pAbs led to development of melanoma cell killing ADCs against LGR6, TRPM1, ASAP1, and MUC18, among others. MUC18 was overexpressed in both tumor cells and tumor-infiltrating blood vessels across major melanoma subtypes, making it a potential dual-compartment and universal melanoma therapeutic target. AMT-253, an MUC18-directed ADC based on topoisomerase I inhibitor exatecan and a self-immolative T moiety, had a higher therapeutic index compared with its microtubule inhibitor-based counterpart and favorable pharmacokinetics and tolerability in monkeys. AMT-253 exhibited MUC18-specific cytotoxicity through DNA damage and apoptosis and a strong bystander killing effect, leading to potent antitumor activities against melanoma cell line and patient-derived xenograft models. Tumor vasculature targeting by a mouse MUC18-specific antibody-T1000-exatecan conjugate inhibited tumor growth in human melanoma xenografts. Combination therapy of AMT-253 with an antiangiogenic agent generated higher efficacy than single agent in a mucosal melanoma model. Beyond melanoma, AMT-253 was also efficacious in a wide range of MUC18-expressing solid tumors. Efficient target/antibody discovery in combination with the T moiety-exatecan linker-payload exemplified here may facilitate discovery of new ADC to improve cancer treatment. SIGNIFICANCE Discovery of melanoma-targeting antibodies using a proteome-scale array and use of a cutting-edge linker-payload system led to development of a MUC18-targeting antibody-exatecan conjugate with clinical potential for treating major melanoma subtypes.
Collapse
Affiliation(s)
- Jing Shi
- Multitude Therapeutics, Shanghai, China
| | - Tao Jiao
- Department of Renal Cancer and Melanoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Qian Guo
- Department of Renal Cancer and Melanoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Weining Weng
- Multitude Therapeutics, Shanghai, China
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Linjie Ma
- Multitude Therapeutics, Shanghai, China
| | | | | | | | | | | | | | | | - Yanfang Tang
- Multitude Therapeutics, Shanghai, China
- Abmart, Shanghai, China
| | - Wenhao Hu
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Tao Meng
- MabCare Therapeutics, Shanghai, China
- HySlink Therapeutics, Shanghai, China
| | | | - Jun Guo
- Department of Renal Cancer and Melanoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Yan Kong
- Department of Renal Cancer and Melanoma, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Xun Meng
- Multitude Therapeutics, Shanghai, China
- Abmart, Shanghai, China
| |
Collapse
|
5
|
Jong YI, Harmon SK, O'Malley KL. GPCR
Signaling from Intracellular Membranes. GPCRS AS THERAPEUTIC TARGETS 2022:216-298. [DOI: 10.1002/9781119564782.ch8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
6
|
Zeng Y, Du X, Yao X, Qiu Y, Jiang W, Shen J, Li L, Liu X. Mechanism of cell death of endothelial cells regulated by mechanical forces. J Biomech 2021; 131:110917. [PMID: 34952348 DOI: 10.1016/j.jbiomech.2021.110917] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 12/13/2021] [Accepted: 12/14/2021] [Indexed: 12/26/2022]
Abstract
Cell death of endothelial cells (ECs) is a common devastating consequence of various vascular-related diseases. Atherosclerosis, hypertension, sepsis, diabetes, cerebral ischemia and cardiac ischemia/reperfusion injury, and chronic kidney disease remain major causes of morbidity and mortality worldwide, in which ECs are constantly subjected to a great amount of dynamic changed mechanical forces including shear stress, extracellular matrix stiffness, mechanical stretch and microgravity. A thorough understanding of the regulatory mechanisms by which the mechanical forces controlled the cell deaths including apoptosis, autophagy, and pyroptosis is crucial for the development of new therapeutic strategies. In the present review, experimental and clinical data highlight that nutrient depletion, oxidative stress, tumor necrosis factor-α, high glucose, lipopolysaccharide, and homocysteine possess cytotoxic effects in many tissues and induce apoptosis of ECs, and that sphingosine-1-phosphate protects ECs. Nevertheless, EC apoptosis in the context of those artificial microenvironments could be enhanced, reduced or even reversed along with the alteration of patterns of shear stress. An appropriate level of autophagy diminishes EC apoptosis to some extent, in addition to supporting cell survival upon microenvironment challenges. The intervention of pyroptosis showed a profound effect on atherosclerosis. Further cell and animal studies are required to ascertain whether the alterations in the levels of cell deaths and their associated regulatory mechanisms happen at local lesion sites with considerable mechanical force changes, for preventing senescence and cell deaths in the vascular-related diseases.
Collapse
Affiliation(s)
- Ye Zeng
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China.
| | - Xiaoqiang Du
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xinghong Yao
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yan Qiu
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China
| | - Wenli Jiang
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China
| | - Junyi Shen
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China
| | - Liang Li
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xiaoheng Liu
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
7
|
Minimal contribution of IP 3R2 in cardiac differentiation and derived ventricular-like myocytes from human embryonic stem cells. Acta Pharmacol Sin 2020; 41:1576-1586. [PMID: 33037404 DOI: 10.1038/s41401-020-00528-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 09/03/2020] [Indexed: 02/06/2023]
Abstract
Type 2 inositol 1,4,5-trisphosphate receptor (IP3R2) regulates the intracellular Ca2+ release from endoplasmic reticulum in human embryonic stem cells (hESCs), cardiovascular progenitor cells (CVPCs), and mammalian cardiomyocytes. However, the role of IP3R2 in human cardiac development is unknown and its function in mammalian cardiomyocytes is controversial. hESC-derived cardiomyocytes have unique merits in disease modeling, cell therapy, and drug screening. Therefore, understanding the role of IP3R2 in the generation and function of human cardiomyocytes would be valuable for the application of hESC-derived cardiomyocytes. In the current study, we investigated the role of IP3R2 in the differentiation of hESCs to cardiomyocytes and in the hESC-derived cardiomyocytes. By using IP3R2 knockout (IP3R2KO) hESCs, we showed that IP3R2KO did not affect the self-renewal of hESCs as well as the differentiation ability of hESCs into CVPCs and cardiomyocytes. Furthermore, we demonstrated the ventricular-like myocyte characteristics of hESC-derived cardiomyocytes. Under the α1-adrenergic stimulation by phenylephrine (10 μmol/L), the amplitude and maximum rate of depolarization of action potential (AP) were slightly affected in the IP3R2KO hESC-derived cardiomyocytes at differentiation day 90, whereas the other parameters of APs and the Ca2+ transients did not show significant changes compared with these in the wide-type ones. These results demonstrate that IP3R2 has minimal contribution to the differentiation and function of human cardiomyocytes derived from hESCs, thus provide the new knowledge to the function of IP3R2 in the generation of human cardiac lineage cells and in the early cardiomyocytes.
Collapse
|
8
|
Stridh S, Palm F, Takahashi T, Ikegami-Kawai M, Friederich-Persson M, Hansell P. Hyaluronan Production by Renomedullary Interstitial Cells: Influence of Endothelin, Angiotensin II and Vasopressin. Int J Mol Sci 2017; 18:ijms18122701. [PMID: 29236055 PMCID: PMC5751302 DOI: 10.3390/ijms18122701] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 12/05/2017] [Accepted: 12/10/2017] [Indexed: 11/17/2022] Open
Abstract
The content of hyaluronan (HA) in the interstitium of the renal medulla changes in relation to body hydration status. We investigated if hormones of central importance for body fluid homeostasis affect HA production by renomedullary interstitial cells in culture (RMICs). Simultaneous treatment with vasopressin and angiotensin II (Ang II) reduced HA by 69%. No change occurred in the mRNA expressions of hyaluronan synthase 2 (HAS2) or hyaluronidases (Hyals), while Hyal activity in the supernatant increased by 67% and CD44 expression reduced by 42%. The autocoid endothelin (ET-1) at low concentrations (10−10 and 10−8 M) increased HA 3-fold. On the contrary, at a high concentration (10−6 M) ET-1 reduced HA by 47%. The ET-A receptor antagonist BQ123 not only reversed the reducing effect of high ET-1 on HA, but elevated it to the same level as low concentration ET-1, suggesting separate regulating roles for ET-A and ET-B receptors. This was corroborated by the addition of ET-B receptor antagonist BQ788 to low concentration ET-1, which abolished the HA increase. HAS2 and Hyal2 mRNA did not alter, while Hyal1 mRNA was increased at all ET-1 concentrations tested. Hyal activity was elevated the most by high ET-1 concentration, and blockade of ET-A receptors by BQ123 prevented about 30% of this response. The present study demonstrates an important regulatory influence of hormones involved in body fluid balance on HA handling by RMICs, thereby supporting the concept of a dynamic involvement of interstitial HA in renal fluid handling.
Collapse
Affiliation(s)
- Sara Stridh
- Department of Medical Cell Biology, Uppsala University, Biomedical Center, SE-75123 Uppsala, Sweden; (S.S.); (F.P.); (M.F.-P.)
- Department of Health Sciences, Red Cross University College, SE-14152 Stockholm, Sweden
| | - Fredrik Palm
- Department of Medical Cell Biology, Uppsala University, Biomedical Center, SE-75123 Uppsala, Sweden; (S.S.); (F.P.); (M.F.-P.)
| | - Tomoko Takahashi
- Faculty of Pharmaceutical Sciences, Hoshi University, Tokyo 142-8501, Japan; (T.T.); (M.I.-K.)
| | - Mayumi Ikegami-Kawai
- Faculty of Pharmaceutical Sciences, Hoshi University, Tokyo 142-8501, Japan; (T.T.); (M.I.-K.)
| | - Malou Friederich-Persson
- Department of Medical Cell Biology, Uppsala University, Biomedical Center, SE-75123 Uppsala, Sweden; (S.S.); (F.P.); (M.F.-P.)
| | - Peter Hansell
- Department of Medical Cell Biology, Uppsala University, Biomedical Center, SE-75123 Uppsala, Sweden; (S.S.); (F.P.); (M.F.-P.)
- Correspondence: ; Tel.: +46-184-714-130
| |
Collapse
|
9
|
SCF-KIT signaling induces endothelin-3 synthesis and secretion: Thereby activates and regulates endothelin-B-receptor for generating temporally- and spatially-precise nitric oxide to modulate SCF- and or KIT-expressing cell functions. PLoS One 2017; 12:e0184154. [PMID: 28880927 PMCID: PMC5589172 DOI: 10.1371/journal.pone.0184154] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 08/19/2017] [Indexed: 01/11/2023] Open
Abstract
We demonstrate that SCF-KIT signaling induces synthesis and secretion of endothelin-3 (ET3) in human umbilical vein endothelial cells and melanoma cells in vitro, gastrointestinal stromal tumors, human sun-exposed skin, and myenteric plexus of human colon post-fasting in vivo. This is the first report of a physiological mechanism of ET3 induction. Integrating our finding with supporting data from literature leads us to discover a previously unreported pathway of nitric oxide (NO) generation derived from physiological endothelial NO synthase (eNOS) or neuronal NOS (nNOS) activation (referred to as the KIT-ET3-NO pathway). It involves: (1) SCF-expressing cells communicate with neighboring KIT-expressing cells directly or indirectly (cleaved soluble SCF). (2) SCF-KIT signaling induces timely local ET3 synthesis and secretion. (3) ET3 binds to ETBR on both sides of intercellular space. (4) ET3-binding-initiated-ETBR activation increases cytosolic Ca2+, activates cell-specific eNOS or nNOS. (5) Temporally- and spatially-precise NO generation. NO diffuses into neighboring cells, thus acts in both SCF- and KIT-expressing cells. (6) NO modulates diverse cell-specific functions by NO/cGMP pathway, controlling transcriptional factors, or other mechanisms. We demonstrate the critical physiological role of the KIT-ET3-NO pathway in fulfilling high demand (exceeding basal level) of endothelium-dependent NO generation for coping with atherosclerosis, pregnancy, and aging. The KIT-ET3-NO pathway most likely also play critical roles in other cell functions that involve dual requirement of SCF-KIT signaling and NO. New strategies (e.g. enhancing the KIT-ET3-NO pathway) to harness the benefit of endogenous eNOS and nNOS activation and precise NO generation for correcting pathophysiology and restoring functions warrant investigation.
Collapse
|
10
|
Brailoiu GC, Brailoiu E. Modulation of Calcium Entry by the Endo-lysosomal System. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 898:423-47. [PMID: 27161239 DOI: 10.1007/978-3-319-26974-0_18] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Endo-lysosomes are acidic organelles that besides the role in macromolecules degradation, act as intracellular Ca(2+) stores. Nicotinic acid adenine dinucleotide phosphate (NAADP), the most potent Ca(2+)-mobilizing second messenger, produced in response to agonist stimulation, activates Ca(2+)-releasing channels on endo-lysosomes and modulates a variety of cellular functions. NAADP-evoked signals are amplified by Ca(2+) release from endoplasmic reticulum, via the recruitment of inositol 1,4,5-trisphosphate and/or ryanodine receptors through a Ca(2+)-induced Ca(2+)- release (CICR) mechanism. The endo-lysosomal Ca(2+) channels activated by NAADP were recently identified as the two-pore channels (TPCs). In addition to TPCs, endo-lysosomes express another distinct family of Ca(2+)- permeable channels, namely the transient receptor potential mucolipin (TRPML) channels, functionally distinct from TPCs. TPCs belong to the voltage-gated channels, resembling voltage-gated Na(+) and Ca(2+) channels. TPCs have important roles in vesicular fusion and trafficking, in triggering a global Ca(2+) signal and in modulation of the membrane excitability. Depletion of acidic Ca(2+) stores has been shown to activate store-operated Ca(2+) entry in human platelets and mouse pancreatic β-cells. In human platelets, Ca(2+) influx in response to acidic stores depletion is facilitated by the tubulin-cytoskeleton and occurs through non-selective cation channels and transient receptor potential canonical (TRPC) channels. Emerging evidence indicates that activation of intracellular receptors, situated on endo-lysosomes, elicits canonical and non-canonical signaling mechanisms that involve CICR and activation of non-selective cation channels in plasma membrane. The ability of endo-lysosomal Ca(2+) stores to modulate the Ca(2+) release from other organelles and the Ca(2+) entry increases the diversity and complexity of cellular signaling mechanisms.
Collapse
Affiliation(s)
- G Cristina Brailoiu
- Department of Pharmaceutical Sciences, Jefferson School of Pharmacy, Thomas Jefferson University, 901 Walnut St, Rm 916, Philadelphia, PA, 19107, USA.
| | - Eugen Brailoiu
- Center for Substance Abuse Research, Temple University School of Medicine, 3500 N. Broad Street, Room 848, Philadelphia, PA, 19140, USA
| |
Collapse
|
11
|
Detection of autophagy in Hirschsprung's disease: implication for its role in aganglionosis. Neuroreport 2016; 26:1044-50. [PMID: 26509546 DOI: 10.1097/wnr.0000000000000465] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Hirschsprung's disease (HD) is a common congenital gastrointestinal malformation, characterized by the lack of ganglion cells from the distal rectum to the proximal bowel, but the pathogenesis is not well understood. This paper evaluates the effects of autophagy in HD. Using electron microscopy, the autophagosomes were detected in three segments: narrow segment (NS), transitional segment (TS), and dilated segment (DS). Typical autophagosome structures are found in the Auerbach plexus of both NS and TS. Real-time PCR results showed that Beclin1 (NS vs. TS, P<0.01) and LC3 (NS vs. TS, P<0.05) mRNA were the highest in the NS, but p75 (NS vs. TS, P<0.01) was the highest in the DS. Correlation analysis results showed a positive correlation between Beclin1 and LC3 mRNA levels (R=0.736, P=0.000), whereas inverse correlations were found between p75 and Beclin1/LC3 mRNA levels (p75 vs. Beclin1: R=-0.714, P=0.000; p75 vs. LC3: R=-0.619, P=0.000). Immunohistochemistry analyses indicated a consistent result with mRNA levels, by increased Beclin1-positive and LC3-positive neurons, but reduced p75-positive neurons in the Auerbach plexus of TS compared with DS. These findings indicated that autophagy exists in the bowel of patients with HD. On the basis of the detection of the highest expression of the autophagy genes in NS, autophagy may additionally cause the lack of neurons.
Collapse
|
12
|
Park K, Mima A, Li Q, Rask-Madsen C, He P, Mizutani K, Katagiri S, Maeda Y, Wu IH, Khamaisi M, Preil SR, Maddaloni E, Sørensen D, Rasmussen LM, Huang PL, King GL. Insulin decreases atherosclerosis by inducing endothelin receptor B expression. JCI Insight 2016; 1:e86574. [PMID: 27200419 PMCID: PMC4869734 DOI: 10.1172/jci.insight.86574] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Endothelial cell (EC) insulin resistance and dysfunction, caused by diabetes, accelerates atherosclerosis. It is unknown whether specifically enhancing EC-targeted insulin action can decrease atherosclerosis in diabetes. Accordingly, overexpressing insulin receptor substrate-1 (IRS1) in the endothelia of Apoe-/- mice (Irs1/Apoe-/-) increased insulin signaling and function in the aorta. Atherosclerosis was significantly reduced in Irs1/ApoE-/- mice on diet-induced hyperinsulinemia and hyperglycemia. The mechanism of insulin's enhanced antiatherogenic actions in EC was related to remarkable induction of NO action, which increases endothelin receptor B (EDNRB) expression and intracellular [Ca2+]. Using the mice with knockin mutation of eNOS, which had Ser1176 mutated to alanine (AKI), deleting the only known mechanism for insulin to activate eNOS/NO pathway, we observed that IRS1 overexpression in the endothelia of Aki/ApoE-/- mice significantly decreased atherosclerosis. Interestingly, endothelial EDNRB expression was selectively reduced in intima of arteries from diabetic patients and rodents. However, endothelial EDNRB expression was upregulated by insulin via P13K/Akt pathway. Finally EDNRB deletion in EC of Ldlr-/- and Irs1/Ldlr-/- mice decreased NO production and accelerated atherosclerosis, compared with Ldlr-/- mice. Accelerated atherosclerosis in diabetes may be reduced by improving insulin signaling selectively via IRS1/Akt in the EC by inducing EDNRB expression and NO production.
Collapse
Affiliation(s)
- Kyoungmin Park
- Dianne Nunnally Hoppes Laboratory Section of Vascular Cell Biology, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Akira Mima
- Dianne Nunnally Hoppes Laboratory Section of Vascular Cell Biology, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Qian Li
- Dianne Nunnally Hoppes Laboratory Section of Vascular Cell Biology, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Christian Rask-Madsen
- Dianne Nunnally Hoppes Laboratory Section of Vascular Cell Biology, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Pingnian He
- Department of Cellular and Molecular Physiology, Penn State Hershey College of Medicine, Hershey, Pennsylvania, USA
| | - Koji Mizutani
- Dianne Nunnally Hoppes Laboratory Section of Vascular Cell Biology, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Sayaka Katagiri
- Dianne Nunnally Hoppes Laboratory Section of Vascular Cell Biology, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Yasutaka Maeda
- Dianne Nunnally Hoppes Laboratory Section of Vascular Cell Biology, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, USA
| | - I-Hsien Wu
- Dianne Nunnally Hoppes Laboratory Section of Vascular Cell Biology, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Mogher Khamaisi
- Dianne Nunnally Hoppes Laboratory Section of Vascular Cell Biology, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Simone Rordam Preil
- Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, University of Southern Denmark, Odense, Denmark
| | - Ernesto Maddaloni
- Dianne Nunnally Hoppes Laboratory Section of Vascular Cell Biology, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Ditte Sørensen
- Dianne Nunnally Hoppes Laboratory Section of Vascular Cell Biology, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, USA
- Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, University of Southern Denmark, Odense, Denmark
- Danish Diabetes Academy, Odense, Denmark
| | - Lars Melholt Rasmussen
- Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, University of Southern Denmark, Odense, Denmark
| | - Paul L. Huang
- Cardiovascular Research Center, Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| | - George L. King
- Dianne Nunnally Hoppes Laboratory Section of Vascular Cell Biology, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
13
|
Kharitonova M, Iezhitsa I, Zheltova A, Ozerov A, Spasov A, Skalny A. Comparative angioprotective effects of magnesium compounds. J Trace Elem Med Biol 2015; 29:227-34. [PMID: 25127069 DOI: 10.1016/j.jtemb.2014.06.026] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Revised: 06/30/2014] [Accepted: 06/30/2014] [Indexed: 01/10/2023]
Abstract
Magnesium (Mg) deficiency is implicated in the development of numerous disorders of the cardiovascular system. Moreover, the data regarding the efficacy of different magnesium compounds in the correction of impaired functions due to low magnesium intake are often fragmentary and inconsistent. The aim of this study was to compare the effects of the most bioavailable Mg compounds (Mg l-aspartate, Mg N-acetyltaurate, Mg chloride, Mg sulphate and Mg oxybutyrate) on systemic inflammation and endothelial dysfunction in rats fed a low Mg diet for 74 days. A low Mg diet decreased the Mg concentration in the plasma and erythrocytes, which was accompanied by a reduced concentration of eNOs and increased levels of endothelin-1 level in the serum and impaired endothelium-dependent vasodilatation. These effects increased the concentration of proinflammatory molecules, such as VCAM-1, TNF-α, IL-6 and CRP, indicating the development of systemic inflammation and endothelial dysfunction. The increased total NO level, which estimated from the sum of the nitrate and nitrite concentrations in the serum, may also be considered to be a proinflammatory marker. Two weeks of Mg supplementation partially or fully normalised the ability of the vascular wall to effect adequate endothelium-dependent vasodilatation and reversed the levels of most endothelial dysfunction and inflammatory markers (except CRP) to the mean values of the control group. Mg sulphate had the smallest effect on the endothelin-1, TNF-α and VCAM-1 levels. Mg N-acetyltaurate was significantly more effective in restoring the level of eNOS compared to all other studied compounds, except for Mg oxybutyrate. Taken together, the present findings demonstrate that all Mg compounds equally alleviate endothelial dysfunction and inflammation caused by Mg deficiency. Mg sulphate tended to be the least effective compound.
Collapse
Affiliation(s)
- Maria Kharitonova
- Department of Pharmacology, Volgograd State Medical University, Pl. Pavshih Bortsov, 1, Volgograd 400131, Russia; Institute of Pharmacy, Department of Pharmacology and Toxicology, University of Innsbruck, Center for Chemistry and Biomedicine, Innrain 80-82/III, A-6020 Innsbruck, Austria
| | - Igor Iezhitsa
- Department of Pharmacology, Volgograd State Medical University, Pl. Pavshih Bortsov, 1, Volgograd 400131, Russia; Universiti Teknologi MARA, Faculty of Medicine, Sungai Buloh Campus, Jalan Hospital, 47000 Sungai Buloh, Selangor Darul Ehsan, Malaysia; Universiti Teknologi MARA (UiTM), RIG "Molecular Pharmacology and Advanced Therapeutics", Brain and Neuroscience Communities of Research, 40450 Shah Alam, Selangor Darul Ehsan, Malaysia.
| | - Anastasia Zheltova
- Department of Pharmacology, Volgograd State Medical University, Pl. Pavshih Bortsov, 1, Volgograd 400131, Russia; Department of Allergology and Immunology, Volgograd State Medical University, Pl. Pavshih Bortsov, 1, Volgograd 400131, Russia
| | - Alexander Ozerov
- Department for Pharmaceutical and Toxicological Chemistry, Volgograd State Medical University, Pl. Pavshih Bortsov, 1, Volgograd 400131, Russia
| | - Alexander Spasov
- Department of Pharmacology, Volgograd State Medical University, Pl. Pavshih Bortsov, 1, Volgograd 400131, Russia
| | - Anatoly Skalny
- Russian Society of Trace Elements in Medicine, 46 Zemlyanoy Val str., Moscow 105064, Russia; Trace Element - Institute for UNESCO, 7 rue Guillaume Paradin, 69008 Lyon, France
| |
Collapse
|
14
|
Visualization of nitric oxide production by individual platelets during adhesion in flowing blood. Blood 2014; 125:697-705. [PMID: 25480660 DOI: 10.1182/blood-2014-06-579474] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Nitric oxide (NO) exerts vasodilatatory, antiplatelet, antioxidant, and antiproliferative effects. Endothelium-derived NO has been shown to be of crucial importance in cardiovascular protection, whereas evidence that NO is synthesized by platelets and regulates platelet function is still controversial. By using a sensitive and specific fluorescent probe, 4-amino-5-methylamino-2',7'-difluorofluorescein diacetate (DAF-FM), we visualized NO production in individual platelets undergoing adhesion on a collagen substrate under flow conditions. NO production, monitored in real time, was dependent on the shear rates applied, increasing with the raising of the shear rates. Furthermore, NO production increased in the presence of l-arginine (nitric-oxide synthase [NOS] substrate), and it decreased in the presence of L-NG-monomethyl arginine (L-NMMA) (NOS inhibitor) but not of D-NG-monomethyl arginine (D-NMMA) (L-NMMA-inactive enantiomer). Platelet deposition, measured with mepacrine-labeled platelets, was inversely related to NO production. A correlation was evident between Ca(++) elevation and NO production, suggesting that platelet NO formation is triggered by intracytoplasmic Ca(++) elevation. Simultaneous measurement of NO and Ca(++) indicated that NO production in individual platelets is preceded by Ca(++) elevations, with a lag phase of 33 ± 9.5 s. Our studies provide the first direct demonstration of platelet NO production triggered by the interaction with an activating surface under flow and suggest that intraplatelet Ca(++) elevation elicits the production of NO which, in turn, modulates thrombus size.
Collapse
|
15
|
Hockey LN, Kilpatrick BS, Eden ER, Lin-Moshier Y, Brailoiu GC, Brailoiu E, Futter CE, Schapira AH, Marchant JS, Patel S. Dysregulation of lysosomal morphology by pathogenic LRRK2 is corrected by TPC2 inhibition. J Cell Sci 2014; 128:232-8. [PMID: 25416817 PMCID: PMC4294771 DOI: 10.1242/jcs.164152] [Citation(s) in RCA: 132] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Two-pore channels (TPCs) are endolysosomal ion channels implicated in Ca2+ signalling from acidic organelles. The relevance of these ubiquitous proteins for human disease, however, is unclear. Here, we report that lysosomes are enlarged and aggregated in fibroblasts from Parkinson disease patients with the common G2019S mutation in LRRK2. Defects were corrected by molecular silencing of TPC2, pharmacological inhibition of TPC regulators [Rab7, NAADP and PtdIns(3,5)P2] and buffering local Ca2+ increases. NAADP-evoked Ca2+ signals were exaggerated in diseased cells. TPC2 is thus a potential drug target within a pathogenic LRRK2 cascade that disrupts Ca2+-dependent trafficking in Parkinson disease.
Collapse
Affiliation(s)
- Leanne N Hockey
- Department of Cell and Developmental Biology, University College London, Gower Street, London, WC1E 6BT, UK
| | - Bethan S Kilpatrick
- Department of Cell and Developmental Biology, University College London, Gower Street, London, WC1E 6BT, UK
| | - Emily R Eden
- Department of Cell Biology, Institute of Ophthalmology, University College London, London, EC1V 9EL, UK
| | - Yaping Lin-Moshier
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, Minnesota, 55455, USA
| | - G Cristina Brailoiu
- Department of Pharmaceutical Sciences, Thomas Jefferson University, Jefferson School of Pharmacy, Philadelphia, 19107, USA
| | - Eugen Brailoiu
- Department of Pharmacology and Center for Substance Abuse Research, Temple University School of Medicine, Philadelphia, 19140, USA
| | - Clare E Futter
- Department of Cell Biology, Institute of Ophthalmology, University College London, London, EC1V 9EL, UK
| | - Anthony H Schapira
- Department of Clinical Neurosciences, Institute of Neurology, University College London, London, NW3 2PF, UK
| | - Jonathan S Marchant
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, Minnesota, 55455, USA
| | - Sandip Patel
- Department of Cell and Developmental Biology, University College London, Gower Street, London, WC1E 6BT, UK
| |
Collapse
|
16
|
Brailoiu GC, Deliu E, Marcu J, Hoffman NE, Console-Bram L, Zhao P, Madesh M, Abood ME, Brailoiu E. Differential activation of intracellular versus plasmalemmal CB2 cannabinoid receptors. Biochemistry 2014; 53:4990-9. [PMID: 25033246 PMCID: PMC4144709 DOI: 10.1021/bi500632a] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
![]()
The therapeutic and psychoactive
properties of cannabinoids have
long been recognized. The type 2 receptor for cannabinoids (CB2) has emerged as an important therapeutic target in several
pathologies, as it mediates beneficial effects of cannabinoids while
having little if any psychotropic activity. Difficulties associated
with the development of CB2-based therapeutic agents have
been related to its intricate pharmacology, including the species
specificity and functional selectivity of the CB2-initiated
responses. We postulated that a plasmalemmal or subcellular location
of the receptor may contribute to the differential signaling pathways
initiated by its activation. To differentiate between these two, we
used extracellular and intracellular administration of CB2 ligands and concurrent calcium imaging in CB2-expressing
U2OS cells. We found that extracellular administration of anandamide
was ineffective, whereas 2-arachidonoyl glycerol (2-AG) and WIN55,212-2
triggered delayed, CB2-dependent Ca2+ responses
that were Gq protein-mediated. When microinjected, all agonists elicited
fast, transient, and dose-dependent elevations in intracellular Ca2+ concentration upon activation of Gq-coupled CB2 receptors. The CB2 dependency was confirmed by the sensitivity
to AM630, a selective CB2 antagonist, and by the unresponsiveness
of untransfected U2OS cells to 2-AG, anandamide, or WIN55,212-2. Moreover,
we provide functional and morphological evidence that CB2 receptors are localized at the endolysosomes, while their activation
releases Ca2+ from inositol 1,4,5-trisphosphate-sensitive-
and acidic-like Ca2+ stores. Our results support the functionality
of intracellular CB2 receptors and their ability to couple
to Gq and elicit Ca2+ signaling. These findings add further
complexity to CB2 receptor pharmacology and argue for careful
consideration of receptor localization in the development of CB2-based therapeutic agents.
Collapse
Affiliation(s)
- G Cristina Brailoiu
- Department of Pharmaceutical Sciences, Thomas Jefferson University School of Pharmacy , Philadelphia, Pennsylvania 19107, United States
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
De Mey JGR, Vanhoutte PM. End o' the line revisited: moving on from nitric oxide to CGRP. Life Sci 2014; 118:120-8. [PMID: 24747136 DOI: 10.1016/j.lfs.2014.04.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Revised: 04/04/2014] [Accepted: 04/05/2014] [Indexed: 12/24/2022]
Abstract
When endothelin-1(ET-1) was discovered it was hailed as the prototypical endothelium-derived contracting factor (EDCF). However, over the years little evidence emerged convincingly demonstrating that the peptide actually contributes to moment-to-moment changes in vascular tone elicited by endothelial cells. This has been attributed to the profound inhibitory effect of nitric oxide (NO) on both the production (by the endothelium) and the action (on vascular smooth muscle) of ET-1. Hence, the peptide is likely to initiate acute changes in vascular diameter only under extreme conditions of endothelial dysfunction when the NO bioavailability is considerably reduced if not absent. The present essay discusses whether or not this concept should be revised, in particular in view of the potent inhibitory effect exerted by calcitonin gene related peptide (CGRP) released from sensorimotor nerves on vasoconstrictor responses to ET-1.
Collapse
Affiliation(s)
- Jo G R De Mey
- Institute of Molecular Medicine, University of South Denmark, Odense, Denmark; Cardiovascular Research Institute Maastricht, Maastricht, the Netherlands
| | - Paul M Vanhoutte
- Institute of Molecular Medicine, University of South Denmark, Odense, Denmark; Department of Pharmacology and Pharmacy and State Key Laboratory for Pharmaceutical Biotechnology, University of Hong Kong, Hong Kong, China.
| |
Collapse
|
18
|
Zhang J, Yang W, Hu B, Wu W, Fallon MB. Endothelin-1 activation of the endothelin B receptor modulates pulmonary endothelial CX3CL1 and contributes to pulmonary angiogenesis in experimental hepatopulmonary syndrome. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:1706-14. [PMID: 24731444 DOI: 10.1016/j.ajpath.2014.02.027] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Revised: 01/29/2014] [Accepted: 02/11/2014] [Indexed: 02/06/2023]
Abstract
Hepatic production and release of endothelin-1 (ET-1) binding to endothelin B (ETB) receptors, overexpressed in the lung microvasculature, is associated with accumulation of pro-angiogenic monocytes and vascular remodeling in experimental hepatopulmonary syndrome (HPS) after common bile duct ligation (CBDL). We have recently found that lung vascular monocyte adhesion and angiogenesis in HPS involve interaction of endothelial C-X3-C motif ligand 1 (CX3CL1) with monocyte CX3C chemokine receptor 1 (CX3CR1), although whether ET-1/ETB receptor activation influences these events is unknown. Our aim was to define if ET-1/ETB receptor activation modulates CX3CL1/CX3CR1 signaling and lung angiogenesis in experimental HPS. A selective ETB receptor antagonist, BQ788, was given for 2 weeks to 1-week CBDL rats. ET-1 (±BQ788) was given to cultured rat pulmonary microvascular endothelial cells overexpressing ETB receptors. BQ788 treatment significantly decreased lung angiogenesis, monocyte accumulation, and CX3CL1 levels after CBDL. ET-1 treatment significantly induced CX3CL1 production in lung microvascular endothelial cells, which was blocked by inhibitors of Ca(2+) and mitogen-activated protein kinase (MEK)/ERK pathways. ET-1-induced ERK activation was Ca(2+) independent. ET-1 administration also increased endothelial tube formation in vitro, which was inhibited by BQ788 or by blocking Ca(2+) and MEK/ERK activation. CX3CR1 neutralizing antibody partially inhibited ET-1 effects on tube formation. These findings identify a novel mechanistic interaction between the ET-1/ETB receptor axis and CX3CL1/CX3CR1 in mediating pulmonary angiogenesis and vascular monocyte accumulation in experimental HPS.
Collapse
Affiliation(s)
- Junlan Zhang
- Division of Gastroenterology, Hepatology, and Nutrition, the Department of Internal Medicine, The University of Texas Health Science Center at Houston, Houston, Texas
| | - Wenli Yang
- Division of Gastroenterology, Hepatology, and Nutrition, the Department of Internal Medicine, The University of Texas Health Science Center at Houston, Houston, Texas
| | - Bingqian Hu
- Division of Gastroenterology, Hepatology, and Nutrition, the Department of Internal Medicine, The University of Texas Health Science Center at Houston, Houston, Texas
| | - Wei Wu
- Division of Gastroenterology, Hepatology, and Nutrition, the Department of Internal Medicine, The University of Texas Health Science Center at Houston, Houston, Texas
| | - Michael B Fallon
- Division of Gastroenterology, Hepatology, and Nutrition, the Department of Internal Medicine, The University of Texas Health Science Center at Houston, Houston, Texas.
| |
Collapse
|
19
|
Spencer NY, Engelhardt JF. The basic biology of redoxosomes in cytokine-mediated signal transduction and implications for disease-specific therapies. Biochemistry 2014; 53:1551-64. [PMID: 24555469 PMCID: PMC3985689 DOI: 10.1021/bi401719r] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
![]()
Redox
reactions have been established as major biological players
in many cellular signaling pathways. Here we review mechanisms of
redox signaling with an emphasis on redox-active signaling endosomes.
Signals are transduced by relatively few reactive oxygen species (ROS),
through very specific redox modifications of numerous proteins and
enzymes. Although ROS signals are typically associated with cellular
injury, these signaling pathways are also critical for maintaining
cellular health at homeostasis. An important component of ROS signaling
pertains to localization and tightly regulated signal transduction
events within discrete microenvironments of the cell. One major aspect
of this specificity is ROS compartmentalization within membrane-enclosed
organelles such as redoxosomes (redox-active endosomes) and the nuclear
envelope. Among the cellular proteins that produce superoxide are
the NADPH oxidases (NOXes), transmembrane proteins that are implicated
in many types of redox signaling. NOXes produce superoxide on only
one side of a lipid bilayer; as such, their orientation dictates the
compartmentalization of ROS and the local control of signaling events
limited by ROS diffusion and/or movement through channels associated
with the signaling membrane. NOX-dependent ROS signaling pathways
can also be self-regulating, with molecular redox sensors that limit
the local production of ROS required for effective signaling. ROS
regulation of the Rac-GTPase, a required co-activator of many NOXes,
is an example of this type of sensor. A deeper understanding of redox
signaling pathways and the mechanisms that control their specificity
will provide unique therapeutic opportunities for aging, cancer, ischemia-reperfusion
injury, and neurodegenerative diseases.
Collapse
Affiliation(s)
- Netanya Y Spencer
- Department of Anatomy and Cell Biology, The University of Iowa , Iowa City, Iowa 52242-1009, United States
| | | |
Collapse
|
20
|
Deliu E, Brailoiu GC, Eguchi S, Hoffman NE, Rabinowitz JE, Tilley DG, Madesh M, Koch WJ, Brailoiu E. Direct evidence of intracrine angiotensin II signaling in neurons. Am J Physiol Cell Physiol 2014; 306:C736-44. [PMID: 24401846 DOI: 10.1152/ajpcell.00131.2013] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The existence of a local renin-angiotensin system (RAS) in neurons was first postulated 40 years ago. Further studies indicated intraneuronal generation of ANG II. However, the function and signaling mechanisms of intraneuronal ANG II remained elusive. Since ANG II type 1 receptor (AT1R) is the major type of receptor mediating the effects of ANG II, we used intracellular microinjection and concurrent Ca(2+) and voltage imaging to examine the functionality of intracellular AT1R in neurons. We show that intracellular administration of ANG II produces a dose-dependent elevation of cytosolic Ca(2+) concentration ([Ca(2+)]i) in hypothalamic neurons that is sensitive to AT1R antagonism. Endolysosomal, but not Golgi apparatus, disruption prevents the effect of microinjected ANG II on [Ca(2+)]i. Additionally, the ANG II-induced Ca(2+) response is dependent on microautophagy and sensitive to inhibition of PLC or antagonism of inositol 1,4,5-trisphosphate receptors. Furthermore, intracellular application of ANG II produces AT1R-mediated depolarization of hypothalamic neurons, which is dependent on [Ca(2+)]i increase and on cation influx via transient receptor potential canonical channels. In summary, we provide evidence that intracellular ANG II activates endolysosomal AT1Rs in hypothalamic neurons. Our results point to the functionality of a novel intraneuronal angiotensinergic pathway, extending the current understanding of intracrine ANG II signaling.
Collapse
Affiliation(s)
- Elena Deliu
- Department of Pharmacology, Temple University School of Medicine, Philadelphia, Pennsylvania
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Yu J, Deliu E, Zhang XQ, Hoffman NE, Carter RL, Grisanti LA, Brailoiu GC, Madesh M, Cheung JY, Force T, Abood ME, Koch WJ, Tilley DG, Brailoiu E. Differential activation of cultured neonatal cardiomyocytes by plasmalemmal versus intracellular G protein-coupled receptor 55. J Biol Chem 2013; 288:22481-92. [PMID: 23814062 DOI: 10.1074/jbc.m113.456178] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The L-α-lysophosphatidylinositol (LPI)-sensitive receptor GPR55 is coupled to Ca(2+) signaling. Low levels of GPR55 expression in the heart have been reported. Similar to other G protein-coupled receptors involved in cardiac function, GPR55 may be expressed both at the sarcolemma and intracellularly. Thus, to explore the role of GPR55 in cardiomyocytes, we used calcium and voltage imaging and extracellular administration or intracellular microinjection of GPR55 ligands. We provide the first evidence that, in cultured neonatal ventricular myocytes, LPI triggers distinct signaling pathways via GPR55, depending on receptor localization. GPR55 activation at the sarcolemma elicits, on one hand, Ca(2+) entry via L-type Ca(2+) channels and, on the other, inositol 1,4,5-trisphosphate-dependent Ca(2+) release. The latter signal is further amplified by Ca(2+)-induced Ca(2+) release via ryanodine receptors. Conversely, activation of GPR55 at the membrane of intracellular organelles promotes Ca(2+) release from acidic-like Ca(2+) stores via the endolysosomal NAADP-sensitive two-pore channels. This response is similarly enhanced by Ca(2+)-induced Ca(2+) release via ryanodine receptors. Extracellularly applied LPI produces Ca(2+)-independent membrane depolarization, whereas the Ca(2+) signal induced by intracellular microinjection of LPI converges to hyperpolarization of the sarcolemma. Collectively, our findings point to GPR55 as a novel G protein-coupled receptor regulating cardiac function at two cellular sites. This work may serve as a platform for future studies exploring the potential of GPR55 as a therapeutic target in cardiac disorders.
Collapse
Affiliation(s)
- Justine Yu
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania 19140, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|