1
|
Mitsuyama Y, Goda A, Kikuchi H, Takeuchi K, Inami T, Soejima K, Kohno T. Pulmonary Arterial Hypertension Associated With Idiopathic Hypereosinophilic Syndrome: Importance of Eosinophilia Control With Steroid Therapy. CJC Open 2025; 7:123-125. [PMID: 39872656 PMCID: PMC11763537 DOI: 10.1016/j.cjco.2024.09.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 09/15/2024] [Indexed: 01/30/2025] Open
Affiliation(s)
| | - Ayumi Goda
- Department of Cardiovascular Medicine, Kyorin University, Tokyo, Japan
| | - Hanako Kikuchi
- Department of Cardiovascular Medicine, Kyorin University, Tokyo, Japan
| | - Kaori Takeuchi
- Department of Cardiovascular Medicine, Kyorin University, Tokyo, Japan
| | - Takumi Inami
- Department of Cardiovascular Medicine, Kyorin University, Tokyo, Japan
| | - Kyoko Soejima
- Department of Cardiovascular Medicine, Kyorin University, Tokyo, Japan
| | - Takashi Kohno
- Department of Cardiovascular Medicine, Kyorin University, Tokyo, Japan
| |
Collapse
|
2
|
Yamamura A, Fujiwara M, Kawade A, Amano T, Hossain A, Nayeem MJ, Kondo R, Suzuki Y, Inoue Y, Hayashi H, Suzuki S, Sato M, Yamamura H. Corosolic acid attenuates platelet-derived growth factor signaling in macrophages and smooth muscle cells of pulmonary arterial hypertension. Eur J Pharmacol 2024; 973:176564. [PMID: 38614383 DOI: 10.1016/j.ejphar.2024.176564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 03/28/2024] [Accepted: 04/03/2024] [Indexed: 04/15/2024]
Abstract
Pulmonary arterial hypertension (PAH) is a progressive and life-threatening disease that is characterized by vascular remodeling of the pulmonary artery. Pulmonary vascular remodeling is primarily caused by the excessive proliferation and migration of pulmonary arterial smooth muscle cells (PASMCs), which are facilitated by perivascular inflammatory cells including macrophages. Corosolic acid (CRA) is a natural pentacyclic triterpenoid that exerts anti-inflammatory effects. In the present study, the effects of CRA on the viability of macrophages were examined using monocrotaline (MCT)-induced PAH rats and human monocyte-derived macrophages. Although we previously reported that CRA inhibited signal transducer and activator of transcription 3 (STAT3) signaling and ameliorated pulmonary vascular remodeling in PAH, the inhibitory mechanism remains unclear. Therefore, the underlying mechanisms were investigated using PASMCs from idiopathic PAH (IPAH) patients. In MCT-PAH rats, CRA inhibited the accumulation of macrophages around remodeled pulmonary arteries. CRA reduced the viability of human monocyte-derived macrophages. In IPAH-PASMCs, CRA attenuated cell proliferation and migration facilitated by platelet-derived growth factor (PDGF)-BB released from macrophages and PASMCs. CRA also downregulated the expression of PDGF receptor β and its signaling pathways, STAT3 and nuclear factor-κB (NF-κB). In addition, CRA attenuated the phosphorylation of PDGF receptor β and STAT3 following the PDGF-BB simulation. The expression and phosphorylation levels of PDGF receptor β after the PDGF-BB stimulation were reduced by the small interfering RNA knockdown of NF-κB, but not STAT3, in IPAH-PASMCs. In conclusion, CRA attenuated the PDGF-PDGF receptor β-STAT3 and PDGF-PDGF receptor β-NF-κB signaling axis in macrophages and PASMCs, and thus, ameliorated pulmonary vascular remodeling in PAH.
Collapse
Affiliation(s)
- Aya Yamamura
- Department of Physiology, Aichi Medical University, Nagakute, Aichi, Japan.
| | - Moe Fujiwara
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Aichi, Japan
| | - Akiko Kawade
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Aichi, Japan
| | - Taiki Amano
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Aichi, Japan
| | - Alamgir Hossain
- Department of Physiology, Aichi Medical University, Nagakute, Aichi, Japan
| | - Md Junayed Nayeem
- Department of Physiology, Aichi Medical University, Nagakute, Aichi, Japan
| | - Rubii Kondo
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Aichi, Japan
| | - Yoshiaki Suzuki
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Aichi, Japan
| | - Yasumichi Inoue
- Department of Cell Signaling, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Aichi, Japan
| | - Hidetoshi Hayashi
- Department of Cell Signaling, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Aichi, Japan
| | - Susumu Suzuki
- Research Creation Support Center, Aichi Medical University, Nagakute, Aichi, Japan
| | - Motohiko Sato
- Department of Physiology, Aichi Medical University, Nagakute, Aichi, Japan
| | - Hisao Yamamura
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Aichi, Japan.
| |
Collapse
|
3
|
Sulforaphane alleviated vascular remodeling in hypoxic pulmonary hypertension via inhibiting inflammation and oxidative stress. J Nutr Biochem 2023; 111:109182. [PMID: 36220525 DOI: 10.1016/j.jnutbio.2022.109182] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 08/29/2022] [Accepted: 09/02/2022] [Indexed: 11/05/2022]
Abstract
Hypoxic pulmonary hypertension (HPH) is a cardiopulmonary disease featured by pulmonary vascular remodeling, which is due to abnormal proliferation of pulmonary artery smooth muscle cells (PASMCs) and dysfunction of endothelial cells (ECs). Sulforaphane (SFN) is a natural isothiocyanate extracted from cruciferous vegetables with promising anti-inflammatory and anti-oxidative activities. This study aimed to explore the effect and mechanism of SFN on HPH. Male mice were exposed to persistent chronic hypoxia for 4 weeks to induce HPH. The results demonstrated that SFN repressed the increased right ventricular systolic pressure (RVSP) and attenuated the right ventricular hypertrophy and pulmonary arteries remodeling in HPH mice. In particular, after SFN treatment, the CD68 positive cells in lung sections were reduced; TNF-α and IL-6 levels in lungs and serum declined; activation of NF-κB in PASMCs was inhibited in response to hypoxia. Besides, SFN enhanced the superoxide dismutase (SOD) activity in serum, SOD2 expression, total glutathione levels, and GSH/GSSG ratio in PASMCs, along with a decrease in malondialdehyde (MDA) contents in serum and ROS production in PASMCs after hypoxia exposure. Notably, SFN, as an Nrf2 activator, reversed the reduction in Nrf2 expression in hypoxic PASMCs. In vitro, SFN treatment inhibited hyperproliferation and promoted apoptosis of PASMCs under hypoxia conditions. SFN also prevented the apoptosis of pulmonary microvascular ECs caused by hypoxia. Therefore, these data suggested that SFN could significantly restrain the inflammation and oxidative stress, thereby inhibiting PASMCs proliferation, promoting PASMCs apoptosis, and reversing hypoxia injury in ECs to improve pulmonary vascular remodeling.
Collapse
|
4
|
Wang J, Wang L, Chen X, Liang ML, Wei DH, Cao W, Zhang J. Cigarette smoke extract stimulates human pulmonary artery smooth muscle cell proliferation: Role of inflammation and oxidative stress. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2022; 25:755-761. [PMID: 35949310 PMCID: PMC9320202 DOI: 10.22038/ijbms.2022.64170.14133] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 06/08/2022] [Indexed: 11/12/2022]
Abstract
Objectives Cigarette smoke may play a direct role in proliferation of human pulmonary artery smooth muscle cells (HPASMCs). However, the mechanism involved and the effect of interventions remain unclear. We aimed to evaluate the effect of cigarette smoke extract (CSE) on HPASMCs, explore the role of inflammation and oxidative stress, and the effects of Tempol and PDTC in this process. Materials and Methods HPASMCs were subjected to normal control (NC), CSE, CSE+Tempol (CSE+T), and CSE+PDTC (CSE+P) groups. Proliferation of HPASMCs was measured by CCK-8 and Western blot. TNF-α, IL-6, MDA, and SOD levels were determined by ELISA and commercial kits. Nuclear translocation of NF-κB p65 was evaluated by western blot. Results 1%, 2.5%, and 5% CSE all promoted proliferation of HPASMCs, and effect of 1% CSE was the most significant, however, 7.5% and 10% CSE inhibited viability of cells (all P<0.05). Compared with the NC group, TNF-α, IL-6, and MDA levels increased, SOD activity decreased (all P<0.05), and NF-κB p65 expression in nuclei increased (P=0.04) in the CSE group. Tempol and PDTC inhibited the proliferation of HPASMCs induced by CSE (all P<0.05). And compared with the CSE group, TNF-α, IL-6, and MDA levels in CSE+T and CSE+P groups decreased, while SOD activity increased (all P<0.05). Tempol reduced the expression of NF-κB p65 in nuclei but did not achieve a significant difference (P=0.08). PDTC inhibited the nuclear translocation of NF-κB p65 (P=0.03). Conclusion CSE stimulates HPASMCs proliferation in a certain concentration range. The CSE-induced proliferation of HPASMCs involved excessive inflammatory response and oxidative stress. Tempol and PDTC attenuate these effects of CSE on HPASMCs.
Collapse
Affiliation(s)
- Juan Wang
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin 300052, China,These authors contributed eqully to this work
| | - Le Wang
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin 300052, China,These authors contributed eqully to this work
| | - Xing Chen
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin 300052, China,These authors contributed eqully to this work
| | - Mao-Li Liang
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Dong-Hui Wei
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Wei Cao
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin 300052, China,Corresponding authors: Jie Cao. Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin 300052, China. Tel: 022-60361612; Fax: 022-60361612; ; Jing Zhang. Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin 300052, China. Tel: 022-60361612; Fax: 022-60361612;
| | - Jing Zhang
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin 300052, China,Corresponding authors: Jie Cao. Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin 300052, China. Tel: 022-60361612; Fax: 022-60361612; ; Jing Zhang. Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin 300052, China. Tel: 022-60361612; Fax: 022-60361612;
| |
Collapse
|
5
|
Wang Y, Li N, Wang Y, Zheng G, An J, Liu C, Wang Y, Liu Q. NF-κB/p65 Competes With Peroxisome Proliferator-Activated Receptor Gamma for Transient Receptor Potential Channel 6 in Hypoxia-Induced Human Pulmonary Arterial Smooth Muscle Cells. Front Cell Dev Biol 2021; 9:656625. [PMID: 34950652 PMCID: PMC8688744 DOI: 10.3389/fcell.2021.656625] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 09/20/2021] [Indexed: 11/13/2022] Open
Abstract
Objective: Peroxisome proliferator-activated receptor gamma (PPARγ) has an anti-proliferation effect on pulmonary arterial smooth muscle cells (PASMCs) via the transient receptor potential channel (TRPC) and protects against pulmonary artery hypertension (PAH), whereas nuclear factor-kappa B (NF-κB) has pro-proliferation and pro-inflammation effects, which contributes to PAH. However, the association between them in PAH pathology remains unclear. Therefore, this study aimed to investigate this association and the mechanisms underlying TRPC1/6 signaling-mediated PAH. Methods: Human pulmonary arterial smooth muscle cells (hPASMCs) were transfected with p65 overexpressing (pcDNA-p65) and interfering plasmids (shp65) and incubated in normal and hypoxic conditions (4% O2 and 72 h). The effects of hypoxia and p65 expression on cell proliferation, invasion, apoptosis, [Ca2+]i, PPARγ, and TRPC1/6 expression were determined using Cell Counting Kit-8 (CCK-8), Transwell, Annexin V/PI, Fura-2/AM, and western blotting, respectively. In addition, the binding of p65 or PPARγ proteins to the TRPC6 promoter was validated using a dual-luciferase report assay, chromatin-immunoprecipitation-polymerase chain reaction (ChIP-PCR), and electrophoretic mobility shift assay (EMSA). Results: Hypoxia inhibited hPASMC apoptosis and promoted cell proliferation and invasion. Furthermore, it increased [Ca2+]i and the expression of TRPC1/6, p65, and Bcl-2 proteins. Moreover, pcDNA-p65 had similar effects on hypoxia treatment by increasing TRPC1/6 expression, [Ca2+]i, hPASMC proliferation, and invasion. The dual-luciferase report and ChIP-PCR assays revealed three p65 binding sites and two PPARγ binding sites on the promoter region of TRPC6. In addition, hypoxia treatment and shPPARγ promoted the binding of p65 to the TRPC6 promoter, whereas shp65 promoted the binding of PPARγ to the TRPC6 promoter. Conclusion: Competitive binding of NF-κB p65 and PPARγ to TRPC6 produced an anti-PAH effect.
Collapse
Affiliation(s)
- Yan Wang
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Naijian Li
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yingfeng Wang
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital of Southern Medical University, Guangzhou, China
- Department of Guangdong Provincial Key Laboratory of Shock and Microcirculation, Guangzhou, China
- *Correspondence: Yingfeng Wang,
| | - Guobing Zheng
- Prenatal Diagnosis Unit, Boai Hospital of Zhongshan, Zhongshan, China
| | - Jing An
- Department of Academic Research Office, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Chang Liu
- Department of Scientific Research Center, Southern Medical University, Guangzhou, China
| | - Yajie Wang
- Dermatology Hospital of Southern Medical University, Guangzhou, China
- Southern Medical University Institute for Global Health and Sexually Transmitted Diseases, Guangzhou, China
| | - Qicai Liu
- Department of Cardiology, Laboratory of Heart Center, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| |
Collapse
|
6
|
Jain PP, Lai N, Xiong M, Chen J, Babicheva A, Zhao T, Parmisano S, Zhao M, Paquin C, Matti M, Powers R, Balistrieri A, Kim NH, Valdez-Jasso D, Thistlethwaite PA, Shyy JYJ, Wang J, Garcia JGN, Makino A, Yuan JXJ. TRPC6, a therapeutic target for pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol 2021; 321:L1161-L1182. [PMID: 34704831 PMCID: PMC8715021 DOI: 10.1152/ajplung.00159.2021] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 10/19/2021] [Accepted: 10/20/2021] [Indexed: 12/20/2022] Open
Abstract
Idiopathic pulmonary arterial hypertension (PAH) is a fatal and progressive disease. Sustained vasoconstriction due to pulmonary arterial smooth muscle cell (PASMC) contraction and concentric arterial remodeling due partially to PASMC proliferation are the major causes for increased pulmonary vascular resistance and increased pulmonary arterial pressure in patients with precapillary pulmonary hypertension (PH) including PAH and PH due to respiratory diseases or hypoxemia. We and others observed upregulation of TRPC6 channels in PASMCs from patients with PAH. A rise in cytosolic Ca2+ concentration ([Ca2+]cyt) in PASMC triggers PASMC contraction and vasoconstriction, while Ca2+-dependent activation of PI3K/AKT/mTOR pathway is a pivotal signaling cascade for cell proliferation and gene expression. Despite evidence supporting a pathological role of TRPC6, no selective and orally bioavailable TRPC6 antagonist has yet been developed and tested for treatment of PAH or PH. In this study, we sought to investigate whether block of receptor-operated Ca2+ channels using a nonselective blocker of cation channels, 2-aminoethyl diphenylborinate (2-APB, administered intraperitoneally) and a selective blocker of TRPC6, BI-749327 (administered orally) can reverse established PH in mice. The results from the study show that intrapulmonary application of 2-APB (40 µM) or BI-749327 (3-10 µM) significantly and reversibly inhibited acute alveolar hypoxia-induced pulmonary vasoconstriction. Intraperitoneal injection of 2-APB (1 mg/kg per day) significantly attenuated the development of PH and partially reversed established PH in mice. Oral gavage of BI-749327 (30 mg/kg, every day, for 2 wk) reversed established PH by ∼50% via regression of pulmonary vascular remodeling. Furthermore, 2-APB and BI-749327 both significantly inhibited PDGF- and serum-mediated phosphorylation of AKT and mTOR in PASMC. In summary, the receptor-operated and mechanosensitive TRPC6 channel is a good target for developing novel treatment for PAH/PH. BI-749327, a selective TRPC6 blocker, is potentially a novel and effective drug for treating PAH and PH due to respiratory diseases or hypoxemia.
Collapse
MESH Headings
- Animals
- Boron Compounds/pharmacology
- Calcium Signaling
- Cells, Cultured
- Gene Expression Regulation/drug effects
- Humans
- Hypertension, Pulmonary/drug therapy
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/pathology
- Mice
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Phosphatidylinositol 3-Kinases/genetics
- Phosphatidylinositol 3-Kinases/metabolism
- Pulmonary Artery/drug effects
- Pulmonary Artery/metabolism
- Pulmonary Artery/pathology
- TOR Serine-Threonine Kinases/genetics
- TOR Serine-Threonine Kinases/metabolism
- TRPC6 Cation Channel/antagonists & inhibitors
- TRPC6 Cation Channel/genetics
- TRPC6 Cation Channel/metabolism
- Vasoconstriction
Collapse
Affiliation(s)
- Pritesh P Jain
- Section of Physiology, University of California, San Diego, La Jolla, California
- Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
| | - Ning Lai
- Section of Physiology, University of California, San Diego, La Jolla, California
- State Key Laboratory of Respiratory Medicine and First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Mingmei Xiong
- Section of Physiology, University of California, San Diego, La Jolla, California
- State Key Laboratory of Respiratory Medicine and First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jiyuan Chen
- Section of Physiology, University of California, San Diego, La Jolla, California
- State Key Laboratory of Respiratory Medicine and First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Aleksandra Babicheva
- Section of Physiology, University of California, San Diego, La Jolla, California
- Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
| | - Tengteng Zhao
- Section of Physiology, University of California, San Diego, La Jolla, California
- Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
| | - Sophia Parmisano
- Section of Physiology, University of California, San Diego, La Jolla, California
| | - Manjia Zhao
- Section of Physiology, University of California, San Diego, La Jolla, California
| | - Cole Paquin
- Section of Physiology, University of California, San Diego, La Jolla, California
| | - Moreen Matti
- Section of Physiology, University of California, San Diego, La Jolla, California
| | - Ryan Powers
- Section of Physiology, University of California, San Diego, La Jolla, California
| | - Angela Balistrieri
- Section of Physiology, University of California, San Diego, La Jolla, California
- Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
| | - Nick H Kim
- Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
| | - Daniela Valdez-Jasso
- Department of Bioengineering, University of California, San Diego, La Jolla, California
| | - Patricia A Thistlethwaite
- Division of Cardiothoracic Surgery, Department of Surgery, University of California, San Diego, La Jolla, California
| | - John Y-J Shyy
- Division of Cardiovascular Medicine, University of California, San Diego, La Jolla, California
| | - Jian Wang
- Section of Physiology, University of California, San Diego, La Jolla, California
- State Key Laboratory of Respiratory Medicine and First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Joe G N Garcia
- Department of Medicine, The University of Arizona, Tucson, Arizona
| | - Ayako Makino
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, California
| | - Jason X-J Yuan
- Section of Physiology, University of California, San Diego, La Jolla, California
- Division of Pulmonary, Critical Care and Sleep Medicine, University of California, San Diego, La Jolla, California
| |
Collapse
|
7
|
mTOR Signaling in Pulmonary Vascular Disease: Pathogenic Role and Therapeutic Target. Int J Mol Sci 2021; 22:ijms22042144. [PMID: 33670032 PMCID: PMC7926633 DOI: 10.3390/ijms22042144] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/13/2021] [Accepted: 02/15/2021] [Indexed: 12/16/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a progressive and fatal disease without a cure. The exact pathogenic mechanisms of PAH are complex and poorly understood, yet a number of abnormally expressed genes and regulatory pathways contribute to sustained vasoconstriction and vascular remodeling of the distal pulmonary arteries. Mammalian target of rapamycin (mTOR) is one of the major signaling pathways implicated in regulating cell proliferation, migration, differentiation, and protein synthesis. Here we will describe the canonical mTOR pathway, structural and functional differences between mTOR complexes 1 and 2, as well as the crosstalk with other important signaling cascades in the development of PAH. The pathogenic role of mTOR in pulmonary vascular remodeling and sustained vasoconstriction due to its contribution to proliferation, migration, phenotypic transition, and gene regulation in pulmonary artery smooth muscle and endothelial cells will be discussed. Despite the progress in our elucidation of the etiology and pathogenesis of PAH over the two last decades, there is a lack of effective therapeutic agents to treat PAH patients representing a significant unmet clinical need. In this review, we will explore the possibility and therapeutic potential to use inhibitors of mTOR signaling cascade to treat PAH.
Collapse
|
8
|
Juwita T, H P Pakpahan W, M Puspitasari I, Mekar Saptarini N, Levita J. Anti-inflammatory Activity of <i>Etlingera elatior</i> (Jack) R.M. Smith Flower on Gastric Ulceration-induced Wistar Rats. Pak J Biol Sci 2020; 23:1193-1200. [PMID: 32981250 DOI: 10.3923/pjbs.2020.1193.1200] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND AND OBJECTIVE Etlingera elatior (Jack) R.M. Smith rhizome, which has been traditionally used to reduce stomach discomfort, was reported to possess anti-inflammatory activity, however, there is a lack of such a study on the flower. MATERIALS AND METHODS To investigate the anti-inflammatory activity of the E. elatior flower extract on gastric ulceration-induced Wistar rats. The Wistar rats were divided into 6 groups. Group 1 was the normal control, group 2 was the negative control (Arabic gum suspension 2%), group 3 was the positive control (quercetin), group 4-6 were treated with E. elatior flower extract dose of 500, 1000 and 2000 mg kg-1 of b.wt., respectively. The rats were conditioned to gastric ulceration. The stomach weight, microscopic and macroscopic evaluation of gastric mucosal damage was examined. Subsequently, the nuclear factor-kappaB-p65 (NF-kappaB-p65) expression in the fundus was Western-blotted by employing β-actin and GAPDH as the loading controls. RESULTS Etlingera elatior flower extract dose of 1000 mg kg-1 b.wt., reduces the ulceration index and the infiltration of inflammatory cells. Western blot analysis showed inhibition of NF-kappaB-p65 expression by E. elatior flower extract dose of 1000 mg kg-1 of b.wt. CONCLUSION Etlingera elatior flower might possess anti-inflammatory activity by downregulating the expression of NF-kappaB-p65 in the fundus of gastric ulceration-induced Wistar rats.
Collapse
|
9
|
Xu T, Shao L, Wang A, Liang R, Lin Y, Wang G, Zhao Y, Hu J, Liu S. CD248 as a novel therapeutic target in pulmonary arterial hypertension. Clin Transl Med 2020; 10:e175. [PMID: 32997414 PMCID: PMC7507048 DOI: 10.1002/ctm2.175] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 08/05/2020] [Accepted: 08/31/2020] [Indexed: 12/21/2022] Open
Abstract
Pulmonary vascular remodeling is the most important pathological characteristic of pulmonary arterial hypertension (PAH). No effective treatment for PAH is currently available because the mechanism underlying vascular remodeling is not completely clear. CD248, also known as endosialin, is a transmembrane protein that is highly expressed in pericytes and fibroblasts. Here, we evaluated the role of CD248 in pulmonary vascular remodeling and the processes of PAH pathogenesis. Activation of CD248 in pulmonary artery smooth muscle cells (PASMCs) was found to be proportional to the severity of PAH. CD248 contributed to platelet-derived growth factor-BB (PDGF-BB)-induced PASMC proliferation and migration along with the shift to more synthetic phenotypes. In contrast, treatment with Cd248 siRNA or the anti-CD248 therapeutic antibody (ontuxizumab) significantly inhibited the PDGF signaling pathway, obstructed NF-κB p65-mediated transcription of Nox4, and decreased reactive oxygen species production induced by PDGF-BB in PAMSCs. In addition, knockdown of CD248 alleviated pulmonary vascular remodeling in rat PAH models. This study provides novel insights into the dysfunction of PASMCs leading to pulmonary vascular remodeling, and provides evidence for anti-remodeling treatment for PAH via the immediate targeting of CD248.
Collapse
Affiliation(s)
- Tao Xu
- Life Science InstituteJinzhou Medical UniversityJinzhouP. R. China
| | - Lei Shao
- Department of CardiologyFirst Teaching Hospital of Tianjin University of Traditional Chinese MedicineTianjinP. R. China
| | - Aimei Wang
- Department of PhysiologyJinzhou Medical UniversityJinzhouP. R. China
| | - Rui Liang
- Department of PhysiologyJinzhou Medical UniversityJinzhouP. R. China
| | - Yuhan Lin
- Department of PhysiologyJinzhou Medical UniversityJinzhouP. R. China
| | - Guan Wang
- Life Science InstituteJinzhou Medical UniversityJinzhouP. R. China
| | - Yan Zhao
- Life Science InstituteJinzhou Medical UniversityJinzhouP. R. China
| | - Jing Hu
- Life Science InstituteJinzhou Medical UniversityJinzhouP. R. China
| | - Shuangyue Liu
- Department of PhysiologyJinzhou Medical UniversityJinzhouP. R. China
| |
Collapse
|
10
|
Morii C, Tanaka HY, Izushi Y, Nakao N, Yamamoto M, Matsubara H, Kano MR, Ogawa A. 3D in vitro Model of Vascular Medial Thickening in Pulmonary Arterial Hypertension. Front Bioeng Biotechnol 2020; 8:482. [PMID: 32509756 PMCID: PMC7251161 DOI: 10.3389/fbioe.2020.00482] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 04/27/2020] [Indexed: 12/11/2022] Open
Abstract
In pulmonary arterial hypertension (PAH), excessive proliferation of pulmonary artery smooth muscle cells (PASMCs) causes vascular medial thickening. Medial thickening is a histopathological hallmark of pulmonary vascular remodeling, the central disease process driving PAH progression. Pulmonary vascular remodeling causes stenosis and/or obstruction of small pulmonary arteries. This leads to increased pulmonary vascular resistance, elevated pulmonary arterial pressure, and ultimately right heart failure. To improve the survival of PAH patients, which remains at approximately 60% at 3 years after diagnosis, the development of novel PAH-targeted drugs is desired. To this end, a detailed understanding of the mechanisms underlying excessive PASMC proliferation and the medial thickening that ensues is necessary. However, a lack of in vitro models that recapitulate medial thickening impedes our deeper understanding of the pathogenetic mechanisms involved. In the present study, we applied 3-dimensional (3D) cell culture technology to develop a novel in vitro model of the pulmonary artery medial layer using human PAH patient-derived PASMCs. The addition of platelet-derived growth factor (PDGF)-BB, a mitogen known to promote excessive PASMC proliferation in PAH, resulted in increased thickness of the 3D-PAH media tissues. Conversely, administration of the PDGF receptor inhibitor imatinib or other clinical PAH drugs inhibited this medial thickening-inducing effect of PDGF-BB. Altogether, by using 3D cell culture technology, we report the generation of an in vitro model of medial thickening in PAH, which had hitherto not been successfully modeled in vitro. This model is potentially useful for assessing the ability of candidate PAH drugs to suppress medial thickening.
Collapse
Affiliation(s)
- Chiharu Morii
- Department of Pharmaceutical Biomedicine, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan.,Division of Molecular and Cellular Medicine, Department of Clinical Science, National Hospital Organization Okayama Medical Center, Okayama, Japan
| | - Hiroyoshi Y Tanaka
- Department of Pharmaceutical Biomedicine, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Yasuhisa Izushi
- Division of Molecular and Cellular Medicine, Department of Clinical Science, National Hospital Organization Okayama Medical Center, Okayama, Japan
| | - Natsumi Nakao
- Department of Pharmaceutical Biomedicine, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Masaya Yamamoto
- Department of Materials Processing, Graduate School of Engineering, Tohoku University, Sendai, Japan.,Graduate School of Biomedical Engineering, Tohoku University, Sendai, Japan
| | - Hiromi Matsubara
- Division of Molecular and Cellular Medicine, Department of Clinical Science, National Hospital Organization Okayama Medical Center, Okayama, Japan
| | - Mitsunobu R Kano
- Department of Pharmaceutical Biomedicine, Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan.,Department of Pharmaceutical Biomedicine, Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama, Japan
| | - Aiko Ogawa
- Division of Molecular and Cellular Medicine, Department of Clinical Science, National Hospital Organization Okayama Medical Center, Okayama, Japan
| |
Collapse
|
11
|
Zhang M, Chang Z, Zhang P, Jing Z, Yan L, Feng J, Hu Z, Xu Q, Zhou W, Ma P, Hao Y, Zhou R. Protective effects of 18β-glycyrrhetinic acid on pulmonary arterial hypertension via regulation of Rho A/Rho kinsase pathway. Chem Biol Interact 2019; 311:108749. [DOI: 10.1016/j.cbi.2019.108749] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 06/26/2019] [Accepted: 07/15/2019] [Indexed: 11/28/2022]
|
12
|
Kurosawa R, Satoh K, Kikuchi N, Kikuchi H, Saigusa D, Al-Mamun ME, Siddique MAH, Omura J, Satoh T, Sunamura S, Nogi M, Numano K, Miyata S, Uruno A, Kano K, Matsumoto Y, Doi T, Aoki J, Oshima Y, Yamamoto M, Shimokawa H. Identification of Celastramycin as a Novel Therapeutic Agent for Pulmonary Arterial Hypertension. Circ Res 2019; 125:309-327. [PMID: 31195886 DOI: 10.1161/circresaha.119.315229] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
RATIONALE Pulmonary arterial hypertension (PAH) is characterized by enhanced proliferation of pulmonary artery smooth muscle cells (PASMCs) accompanying increased production of inflammatory factors and adaptation of the mitochondrial metabolism to a hyperproliferative state. However, all the drugs in clinical use target pulmonary vascular dilatation, which may not be effective for patients with advanced PAH. OBJECTIVE We aimed to discover a novel drug for PAH that inhibits PASMC proliferation. METHODS AND RESULTS We screened 5562 compounds from original library using high-throughput screening system to discover compounds which inhibit proliferation of PASMCs from patients with PAH (PAH-PASMCs). We found that celastramycin, a benzoyl pyrrole-type compound originally found in a bacteria extract, inhibited the proliferation of PAH-PASMCs in a dose-dependent manner with relatively small effects on PASMCs from healthy donors. Then, we made 25 analogs of celastramycin and selected the lead compound, which significantly inhibited cell proliferation of PAH-PASMCs and reduced cytosolic reactive oxygen species levels. Mechanistic analysis demonstrated that celastramycin reduced the protein levels of HIF-1α (hypoxia-inducible factor 1α), which impairs aerobic metabolism, and κB (nuclear factor-κB), which induces proinflammatory signals, in PAH-PASMCs, leading to reduced secretion of inflammatory cytokine. Importantly, celastramycin treatment reduced reactive oxygen species levels in PAH-PASMCs with increased protein levels of Nrf2 (nuclear factor erythroid 2-related factor 2), a master regulator of cellular response against oxidative stress. Furthermore, celastramycin treatment improved mitochondrial energy metabolism with recovered mitochondrial network formation in PAH-PASMCs. Moreover, these celastramycin-mediated effects were regulated by ZFC3H1 (zinc finger C3H1 domain-containing protein), a binding partner of celastramycin. Finally, celastramycin treatment ameliorated pulmonary hypertension in 3 experimental animal models, accompanied by reduced inflammatory changes in the lungs. CONCLUSIONS These results indicate that celastramycin ameliorates pulmonary hypertension, reducing excessive proliferation of PAH-PASMCs with less inflammation and reactive oxygen species levels, and recovered mitochondrial energy metabolism. Thus, celastramycin is a novel drug for PAH that targets antiproliferative effects on PAH-PASMCs.
Collapse
Affiliation(s)
- Ryo Kurosawa
- From the Department of Cardiovascular Medicine (R.K., K.S., N.K., E.A.M., M.A.H.S., J.O., T.S., S.S., M.N., K.N., S.M., H.S.), Sendai, Japan.,Japan Society for the Promotion of Science, Tokyo, Japan (R.K.)
| | - Kimio Satoh
- From the Department of Cardiovascular Medicine (R.K., K.S., N.K., E.A.M., M.A.H.S., J.O., T.S., S.S., M.N., K.N., S.M., H.S.), Sendai, Japan
| | - Nobuhiro Kikuchi
- From the Department of Cardiovascular Medicine (R.K., K.S., N.K., E.A.M., M.A.H.S., J.O., T.S., S.S., M.N., K.N., S.M., H.S.), Sendai, Japan
| | - Haruhisa Kikuchi
- Tohoku University Graduate School of Pharmaceutical Sciences, Sendai, Japan (H.K., K.K., Y.M., T.D., J.A., Y.O.)
| | - Daisuke Saigusa
- Department of Integrative Genomics, Tohoku University Tohoku Medical Megabank Organizaition (D.S., A.U., M.Y.), Sendai, Japan.,Department of Medical Biochemistry, Tohoku University Graduate School of Medicine (D.S., A.U., M.Y.), Sendai, Japan
| | - Md Elias Al-Mamun
- From the Department of Cardiovascular Medicine (R.K., K.S., N.K., E.A.M., M.A.H.S., J.O., T.S., S.S., M.N., K.N., S.M., H.S.), Sendai, Japan
| | - Mohammad A H Siddique
- From the Department of Cardiovascular Medicine (R.K., K.S., N.K., E.A.M., M.A.H.S., J.O., T.S., S.S., M.N., K.N., S.M., H.S.), Sendai, Japan
| | - Junichi Omura
- From the Department of Cardiovascular Medicine (R.K., K.S., N.K., E.A.M., M.A.H.S., J.O., T.S., S.S., M.N., K.N., S.M., H.S.), Sendai, Japan
| | - Taijyu Satoh
- From the Department of Cardiovascular Medicine (R.K., K.S., N.K., E.A.M., M.A.H.S., J.O., T.S., S.S., M.N., K.N., S.M., H.S.), Sendai, Japan
| | - Shinichiro Sunamura
- From the Department of Cardiovascular Medicine (R.K., K.S., N.K., E.A.M., M.A.H.S., J.O., T.S., S.S., M.N., K.N., S.M., H.S.), Sendai, Japan
| | - Masamichi Nogi
- From the Department of Cardiovascular Medicine (R.K., K.S., N.K., E.A.M., M.A.H.S., J.O., T.S., S.S., M.N., K.N., S.M., H.S.), Sendai, Japan
| | - Kazuhiko Numano
- From the Department of Cardiovascular Medicine (R.K., K.S., N.K., E.A.M., M.A.H.S., J.O., T.S., S.S., M.N., K.N., S.M., H.S.), Sendai, Japan
| | - Satoshi Miyata
- From the Department of Cardiovascular Medicine (R.K., K.S., N.K., E.A.M., M.A.H.S., J.O., T.S., S.S., M.N., K.N., S.M., H.S.), Sendai, Japan
| | - Akira Uruno
- Department of Integrative Genomics, Tohoku University Tohoku Medical Megabank Organizaition (D.S., A.U., M.Y.), Sendai, Japan.,Department of Medical Biochemistry, Tohoku University Graduate School of Medicine (D.S., A.U., M.Y.), Sendai, Japan
| | - Kuniyuki Kano
- Tohoku University Graduate School of Pharmaceutical Sciences, Sendai, Japan (H.K., K.K., Y.M., T.D., J.A., Y.O.)
| | - Yotaro Matsumoto
- Tohoku University Graduate School of Pharmaceutical Sciences, Sendai, Japan (H.K., K.K., Y.M., T.D., J.A., Y.O.)
| | - Takayuki Doi
- Tohoku University Graduate School of Pharmaceutical Sciences, Sendai, Japan (H.K., K.K., Y.M., T.D., J.A., Y.O.)
| | - Junken Aoki
- Tohoku University Graduate School of Pharmaceutical Sciences, Sendai, Japan (H.K., K.K., Y.M., T.D., J.A., Y.O.)
| | - Yoshiteru Oshima
- Tohoku University Graduate School of Pharmaceutical Sciences, Sendai, Japan (H.K., K.K., Y.M., T.D., J.A., Y.O.)
| | - Masayuki Yamamoto
- Department of Integrative Genomics, Tohoku University Tohoku Medical Megabank Organizaition (D.S., A.U., M.Y.), Sendai, Japan.,Department of Medical Biochemistry, Tohoku University Graduate School of Medicine (D.S., A.U., M.Y.), Sendai, Japan
| | - Hiroaki Shimokawa
- From the Department of Cardiovascular Medicine (R.K., K.S., N.K., E.A.M., M.A.H.S., J.O., T.S., S.S., M.N., K.N., S.M., H.S.), Sendai, Japan
| |
Collapse
|
13
|
Labazi H, Trask AJ. Coronary microvascular disease as an early culprit in the pathophysiology of diabetes and metabolic syndrome. Pharmacol Res 2017; 123:114-121. [PMID: 28700893 DOI: 10.1016/j.phrs.2017.07.004] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 06/19/2017] [Accepted: 07/04/2017] [Indexed: 01/09/2023]
Abstract
Metabolic syndrome (MetS) is a group of cardio-metabolic risk factors that includes obesity, insulin resistance, hypertension, and dyslipidemia; these are also a combination of independent coronary artery disease (CAD) risk factors. Alarmingly, the prevalence of MetS risk factors are increasing and a leading cause for mortality. In the vasculature, complications from MetS and type 2 diabetes (T2D) can be divided into microvascular (retinopathy and nephropathy) and macrovascular (cardiovascular diseases and erectile dysfunction). In addition to vascular and endothelial dysfunction, vascular remodeling and stiffness are also hallmarks of cardiovascular disease (CVD), and well-characterized vascular changes that are observed in the early stages of hypertension, T2D, and obesity [1-3]. In the heart, the link between obstructive atherosclerosis of coronary macrovessels and myocardial ischemia (MI) is well established. However, recent studies show that abnormalities in the coronary microcirculation are associated with functional and structural changes in coronary microvessels (classically defined as being ≤150-200μm internal diameter), which may cause or contribute to MI even in the absence of obstractive CAD. This suggests a prognostic value of an abnormal coronary microcirculation as an early sub-clinical culprit in the pathogenesis and progression of heart disease in T2D and MetS. The aim of this review is to summarize recent studies investigating the coronary microvascular remodeling in an early pre-atherosclerotic phase of MetS and T2D, and to explore potential mechanisms associated with the timing of coronary microvascular remodeling relative to that of the macrovasculature.
Collapse
Affiliation(s)
- Hicham Labazi
- Center for Cardiovascular Research and The Heart Center, The Research Institute at Nationwide Children's Hospital Columbus, OH, United States
| | - Aaron J Trask
- Center for Cardiovascular Research and The Heart Center, The Research Institute at Nationwide Children's Hospital Columbus, OH, United States; Department of Pediatrics, The Ohio State University Columbus, OH, United States.
| |
Collapse
|
14
|
Li Y, Wang Y, Li Y, Qian Z, Zhu L, Yang D. Osthole attenuates pulmonary arterial hypertension in monocrotaline‑treated rats. Mol Med Rep 2017; 16:2823-2829. [PMID: 28677726 DOI: 10.3892/mmr.2017.6876] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 04/19/2017] [Indexed: 11/05/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is an insidious and progressive disease that is triggered by various cardiopulmonary diseases. Inflammation has an important role in the progression of PAH. Osthole (Ost) is a coumarin that has clear anti‑inflammatory properties. The present study aimed to investigate the effects of Ost on PAH, and to explore the mechanism underlying this effect. Using the monocrotaline (MCT)‑induced PAH rat model, the effects of Ost on PAH were investigated. Rats were subcutaneously administered a single dose of MCT (50 mg/kg) to establish the PAH model, followed by daily treatment with Ost (10 or 20 mg/kg) by gavage for 28 days. The mean pulmonary arterial pressure (mPAP) was measured and histological analysis was performed. The results demonstrated that Ost significantly decreased mPAP, and reduced thickening of the pulmonary artery, compared with in rats in the MCT group. To further determine whether the effects of Ost on MCT‑induced PAH were associated with inflammatory responses, the nuclear factor‑κB (NF‑κB) p65 signaling pathway was investigated by western blot analysis. The results demonstrated that Ost increased inhibition of the NF‑κB p65 signaling pathway. In conclusion, the results of the present study demonstrate that Ost may suppress the progression of MCT‑induced PAH in rats, which may be, at least partially, mediated through modulation of the NF‑κB p65 signaling pathway.
Collapse
Affiliation(s)
- Yeli Li
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Yingwan Wang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Yiqi Li
- Department of Pharmacology, Zunyi Medical University, Zhuhai, Guangdong 519041, P.R. China
| | - Zhiqiang Qian
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Ling Zhu
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Danli Yang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| |
Collapse
|
15
|
Cai P, Kovacs L, Dong S, Wu G, Su Y. BMP4 inhibits PDGF-induced proliferation and collagen synthesis via PKA-mediated inhibition of calpain-2 in pulmonary artery smooth muscle cells. Am J Physiol Lung Cell Mol Physiol 2017; 312:L638-L648. [PMID: 28235949 PMCID: PMC5451598 DOI: 10.1152/ajplung.00260.2016] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 02/09/2017] [Accepted: 02/23/2017] [Indexed: 11/22/2022] Open
Abstract
In the present study, we investigated the effect of bone morphogenetic protein 4 (BMP4) on PDGF-induced cell proliferation and collagen synthesis in pulmonary artery smooth muscle cells (PASMCs). Normal human PASMCs were incubated with and without PDGF-BB in the absence and presence of BMP4 for 0.5 to 24 h. The protein levels of collagen-I, p-Smad2/3, p-Smad1/5, and intracellular active TGF-β1, calpain activity, and cell proliferation were then measured. The results showed that BMP4 induced an increase in p-Smad1/5 but had no effect on the protein levels of collagen-I, p-Smad2/3, and intracellular active TGF-β1 and calpain activity in control PASMCs. Nevertheless, BMP4 attenuated increases in cell proliferation and protein levels of collagen-I, p-Smad2/3, and intracellular active TGF-β1 and calpain activity in PASMCs exposed to PDGF-BB. Moreover, BMP4 increased PKA activity and inhibition of PKA prevented the inhibitory effects of BMP4 on PDGF-BB-induced calpain activation in normal PASMCs. The PKA activator forskolin recapitulated the suppressive effect of BMP4 on PDGF-induced calpain activation. Furthermore, BMP4 prevented a PDGF-induced decrease in calpain-2 phosphorylation at serine-369 in normal PASMCs. Finally, BMP4 did not attenuate PDGF-induced increases in cell proliferation, collagen-I protein levels, and calpain activation and did not induce PKA activation and did not prevent a PDGF-induced decrease in calpain-2 phosphorylation at serine-369 in PASMCs from idiopathic pulmonary arterial hypertension (PAH) patients. These data demonstrate that BMP4 inhibits PDGF-induced cell proliferation and collagen synthesis via PKA-mediated inhibition of calpain-2 in normal PASMCs. The inhibitory effects of BMP4 on PDGF-induced cell proliferation, collagen synthesis, and calpain-2 activation are impaired in PASMCs from PAH patients, which may contribute to pulmonary vascular remodeling in PAH.
Collapse
Affiliation(s)
- Pengcheng Cai
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia.,Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Laszlo Kovacs
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Sam Dong
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Guangyu Wu
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Yunchao Su
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia; .,Department of Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia.,Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia.,Research Service, Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia; and
| |
Collapse
|
16
|
Dexamethasone induces apoptosis in pulmonary arterial smooth muscle cells. Respir Res 2015; 16:114. [PMID: 26382031 PMCID: PMC4574531 DOI: 10.1186/s12931-015-0262-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 08/18/2015] [Indexed: 01/30/2023] Open
Abstract
Background Dexamethasone suppressed inflammation and haemodynamic changes in an animal model of pulmonary arterial hypertension (PAH). A major target for dexamethasone actions is NF-κB, which is activated in pulmonary vascular cells and perivascular inflammatory cells in PAH. Reverse remodelling is an important concept in PAH disease therapy, and further to its anti-proliferative effects, we sought to explore whether dexamethasone augments pulmonary arterial smooth muscle cell (PASMC) apoptosis. Methods Analysis of apoptosis markers (caspase 3, in-situ DNA fragmentation) and NF-κB (p65 and phospho-IKK-α/β) activation was performed on lung tissue from rats with monocrotaline (MCT)-induced pulmonary hypertension (PH), before and after day 14–28 treatment with dexamethasone (5 mg/kg/day). PASMC were cultured from this rat PH model and from normal human lung following lung cancer surgery. Following stimulation with TNF-α (10 ng/ml), the effects of dexamethasone (10−8–10−6 M) and IKK2 (NF-κB) inhibition (AS602868, 0–3 μM (0-3×10−6 M) on IL-6 and CXCL8 release and apoptosis was determined by ELISA and by Hoechst staining. NF-κB activation was measured by TransAm assay. Results Dexamethasone treatment of rats with MCT-induced PH in vivo led to PASMC apoptosis as displayed by increased caspase 3 expression and DNA fragmentation. A similar effect was seen in vitro using TNF-α-simulated human and rat PASMC following both dexamethasone and IKK2 inhibition. Increased apoptosis was associated with a reduction in NF-κB activation and in IL-6 and CXCL8 release from PASMC. Conclusions Dexamethasone exerted reverse-remodelling effects by augmenting apoptosis and reversing inflammation in PASMC possibly via inhibition of NF-κB. Future PAH therapies may involve targeting these important inflammatory pathways.
Collapse
|
17
|
Pousada G, Baloira A, Valverde D. Molecular and clinical analysis of TRPC6 and AGTR1 genes in patients with pulmonary arterial hypertension. Orphanet J Rare Dis 2015; 10:1. [PMID: 25603901 PMCID: PMC4307182 DOI: 10.1186/s13023-014-0216-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 12/17/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Pulmonary arterial hypertension (PAH) is a rare and progressive vascular disorder characterized by increased pulmonary vascular resistance and right heart failure. The aim of this study was to analyze 5'UTR region in canonical transient receptor potential isoform 6 (TRPC6) and 3'UTR region in Angiotensin II type I receptor (AGTR1) genes in patients with idiopathic and associated PAH. Correlation among mutations and clinical and functional parameters was further analyzed. METHODS Analysis of TRPC6 and AGTR1 genes was performed by polymerase chain reaction (PCR) and direct sequencing. We used a non-parametric test to determine if significant differences were found between the groups studied and chi-square test to compare clinical and hemodynamic variables among genotypes. RESULTS Fifty five patients and fifty two controls were included in this study. We found statistically significant differences for c.1-361A > T (p = 0.0077), c.1-254C > G (p < 0.0001) and c.1-218C > T (p = 0.0021) in TRPC6 gene and c.1166A > C (p < 0.001) in AGTR1 gene, between patients and controls. Idiopathic PAH patients (IPAH) and controls presented significant differences for all 3 TRPC6 polymorphisms (p = 0.020), (p = 0.002) and (p = 0.008) respectively, and also showed differences for AGTR1 gene (p < 0.001). In associated PAH (APAH) patients we found statistical differences for c.1-254C > G (p < 0.001) and c.1-218C > T (p = 0.001) in TRPC6 gene and c.1166A > C (p = 0.001) in AGTR1 gene. Several clinical and hemodynamic parameters showed significant differences between carriers and non-carriers of these single nucleotide polymorphisms (SNPs). Nineteen patients were carriers of all 3 SNPs in TRPC6 gene and presented a more severe phenotype with differences in mean pulmonary arterial pressure (p = 0.016), systolic pulmonary arterial pressure (p = 0.040), cardiac index (p < 0.001) and 6 minute walking test (p = 0.049). 16 of these patients harbored the SNP in AGTR1 gene. These patients showed differences in age at diagnosis (p = 0.049), mean pulmonary arterial pressure (p = 0.033), cardiac index (p = 0.002) and 6 minute walking test (p = 0.039). CONCLUSIONS PAH is a rare disease with pulmonary vascular remodeling caused in part by a heterogeneous constellation of genetic arrangements. This study seems to suggest that c.1-361A > T, c.1-254C > G and c.1-218C > T polymorphisms in TRPC6 gene and c.1166A > C polymorphism in AGTR1 could have a role in the development of this disease.
Collapse
Affiliation(s)
- Guillermo Pousada
- Department Biochemistry, Genetics and Immunology, Faculty of Biology, University of Vigo, Campus As Lagoas Marcosende S/N, 36310, Vigo, Spain. .,Instituto de Investigación Biomédica de Vigo (IBIV), Vigo, Spain.
| | - Adolfo Baloira
- Complexo Hospitalario Universitario de Pontevedra, Servicio de Neumología, Pontevedra, Spain.
| | - Diana Valverde
- Department Biochemistry, Genetics and Immunology, Faculty of Biology, University of Vigo, Campus As Lagoas Marcosende S/N, 36310, Vigo, Spain. .,Instituto de Investigación Biomédica de Vigo (IBIV), Vigo, Spain.
| |
Collapse
|
18
|
Papamatheakis DG, Blood AB, Kim JH, Wilson SM. Antenatal hypoxia and pulmonary vascular function and remodeling. Curr Vasc Pharmacol 2014; 11:616-40. [PMID: 24063380 DOI: 10.2174/1570161111311050006] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Revised: 06/25/2012] [Accepted: 07/12/2012] [Indexed: 01/02/2023]
Abstract
This review provides evidence that antenatal hypoxia, which represents a significant and worldwide problem, causes prenatal programming of the lung. A general overview of lung development is provided along with some background regarding transcriptional and signaling systems of the lung. The review illustrates that antenatal hypoxic stress can induce a continuum of responses depending on the species examined. Fetuses and newborns of certain species and specific human populations are well acclimated to antenatal hypoxia. However, antenatal hypoxia causes pulmonary vascular disease in fetuses and newborns of most mammalian species and humans. Disease can range from mild pulmonary hypertension, to severe vascular remodeling and dangerous elevations in pressure. The timing, length, and magnitude of the intrauterine hypoxic stress are important to disease development, however there is also a genetic-environmental relationship that is not yet completely understood. Determining the origins of pulmonary vascular remodeling and pulmonary hypertension and their associated effects is a challenging task, but is necessary in order to develop targeted therapies for pulmonary hypertension in the newborn due to antenatal hypoxia that can both treat the symptoms and curtail or reverse disease progression.
Collapse
Affiliation(s)
- Demosthenes G Papamatheakis
- Center for Perinatal Biology, Loma Linda University School of Medicine, 11234 Anderson Street, Loma Linda, 92350 CA, USA.
| | | | | | | |
Collapse
|
19
|
MiR-223 downregulation promotes glomerular endothelial cell activation by upregulating importin α4 and α5 in IgA nephropathy. Kidney Int 2014; 85:624-35. [DOI: 10.1038/ki.2013.469] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2012] [Revised: 09/08/2013] [Accepted: 09/12/2013] [Indexed: 12/14/2022]
|
20
|
Zeng J, Yi B, Wang Z, Ning J, Wang X, Lu K. Effect of annexin A2 on hepatopulmonary syndrome rat serum-induced proliferation of pulmonary arterial smooth muscle cells. Respir Physiol Neurobiol 2013; 185:332-8. [DOI: 10.1016/j.resp.2012.09.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2012] [Revised: 09/16/2012] [Accepted: 09/18/2012] [Indexed: 01/14/2023]
|
21
|
Abstract
INTRODUCTION Pulmonary arterial hypertension (PAH) encompasses a rare potentially lethal group of diseases characterized by vasoconstriction, in situ thrombosis and vascular remodeling. Most of the existing therapies including endothelin receptor antagonists, prostacyclin and derivatives, or phsophodiesterase-5 inhibitors tackle mainly the endothelial dysfunction, leaving the remodeling suboptimally inhibited. This explains the disease progression that occurs even with combined therapies and the need for other therapies able to adequately inhibit the vascular remodeling. AREAS COVERED Platelet-derived growth factor (PDGF) signaling pathway was demonstrated to be involved in the vascular remodeling in PAH, and therefore, it might be a desirable therapeutic target in this setting. This review discusses the pathogenic role of this pathway in PAH and its potential inhibitory approaches, focusing on imatinib as well as on the existing preclinical data on this compound. EXPERT OPINION Preclinical studies demonstrated that PDGF inhibition with receptor antagonists such as imatinib reduces vascular remodeling. Therefore, PDGF might represent a plausible therapeutic target in this disease. However, compounds able to block this pathway via different mechanisms might also become potential PAH therapies.
Collapse
Affiliation(s)
- Sabina Antonela Antoniu
- Grigore T Popa, University of Medicine and Pharmacy Iaşi, Department of Medicine II -Pulmonary Disease, Pulmonary Disease University Hospital, 30 Dr I Cihac Str, 700115 Iasi, Romania.
| |
Collapse
|
22
|
Patel JK, Clifford RL, Deacon K, Knox AJ. Ciclesonide inhibits TNFα- and IL-1β-induced monocyte chemotactic protein-1 (MCP-1/CCL2) secretion from human airway smooth muscle cells. Am J Physiol Lung Cell Mol Physiol 2012; 302:L785-92. [PMID: 22246000 PMCID: PMC3331580 DOI: 10.1152/ajplung.00257.2011] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Monocyte chemotactic protein-1 (MCP-1) is a member of the CC family of cytokines. It has monocyte and lymphocyte chemotactic activity and stimulates histamine release from basophils. MCP-1 is implicated in the pathogenesis of inflammatory diseases, including asthma. The airway smooth muscle (ASM) layer is thickened in asthma, and the growth factors and cytokines secreted by ASM cells play a role in the inflammatory response of the bronchial wall. Glucocorticoids and β2-agonists are first-line drug treatments for asthma. Little is known about the effect of asthma treatments on MCP-1 production from human ASM cells. Here, we determined the effect of ciclesonide (a glucocorticoid) and formoterol (a β2-agonist) on MCP-1 production from human ASM cells. TNFα and IL-1β induced MCP-1 secretion from human ASM cells. Formoterol had no effect on MCP-1 expression, while ciclesonide significantly inhibited IL-1β- and TNFα-induced MCP-1. Furthermore, ciclesonide inhibited IL-1β- and TNFα-induced MCP-1 mRNA and IL-1β- and TNFα-induced MCP-1 promoter and enhancer luciferase reporters. Western blots showed that ciclesonide had no effect on IκB degradation. Finally, ciclesonide inhibited an NF-κB luciferase reporter. Our data show that ciclesonide inhibits IL-1β- and TNFα-induced MCP-1 production from human ASM cells via a transcriptional mechanism involving inhibition of NF-κB binding.
Collapse
Affiliation(s)
- Jamie K Patel
- Division of Respiratory Medicine, Nottingham Respiratory Biomedical Research Unit, University of Nottingham, United Kingdom
| | | | | | | |
Collapse
|
23
|
Ogawa A, Firth AL, Smith KA, Maliakal MV, Yuan JXJ. PDGF enhances store-operated Ca2+ entry by upregulating STIM1/Orai1 via activation of Akt/mTOR in human pulmonary arterial smooth muscle cells. Am J Physiol Cell Physiol 2011; 302:C405-11. [PMID: 22031597 DOI: 10.1152/ajpcell.00337.2011] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Platelet-derived growth factor (PDGF) and its receptor are known to be substantially elevated in lung tissues and pulmonary arterial smooth muscle cells (PASMC) isolated from patients and animals with pulmonary arterial hypertension. PDGF has been shown to phosphorylate and activate Akt and mammalian target of rapamycin (mTOR) in PASMC. In this study, we investigated the role of PDGF-mediated activation of Akt signaling in the regulation of cytosolic Ca(2+) concentration and cell proliferation. PDGF activated the Akt/mTOR pathway and, subsequently, enhanced store-operated Ca(2+) entry (SOCE) and cell proliferation in human PASMC. Inhibition of Akt attenuated the increase in cytosolic Ca(2+) concentration due to both SOCE and PASMC proliferation. This effect correlated with a significant downregulation of stromal interacting molecule (STIM) and Orai, proposed molecular correlates for SOCE in many cell types. The data from this study present a novel pathway for the regulation of Ca(2+) signaling and PASMC proliferation involving activation of Akt in response to upregulated expression of PDGF. Targeting this pathway may lead to the development of a novel therapeutic option for the treatment of pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Aiko Ogawa
- Department of Clinical Science, National Hospital Organization Okayama Medical Center, Japan
| | | | | | | | | |
Collapse
|
24
|
Ogawa A, Nakamura K, Mizoguchi H, Fujii N, Fujio H, Kusano KF, Ohe T, Ito H. Prednisolone ameliorates idiopathic pulmonary arterial hypertension. Am J Respir Crit Care Med 2011; 183:139-40. [PMID: 21193794 DOI: 10.1164/ajrccm.183.1.139] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
25
|
Sakao S, Tatsumi K, Voelkel NF. Reversible or irreversible remodeling in pulmonary arterial hypertension. Am J Respir Cell Mol Biol 2010; 43:629-34. [PMID: 20008280 PMCID: PMC2993084 DOI: 10.1165/rcmb.2009-0389tr] [Citation(s) in RCA: 117] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2009] [Accepted: 11/21/2009] [Indexed: 12/15/2022] Open
Abstract
Vascular remodeling is an important pathological feature of pulmonary arterial hypertension (PAH), which leads to increased pulmonary vascular resistance, with marked proliferation of pulmonary artery smooth muscle cells (SMC) and/or endothelial cells (EC). Successful treatment of experimental PAH with a platelet-derived growth factor (PDGF) receptor tyrosine kinase inhibitor offers the perspective of "reverse remodeling" (i.e., the regression of established pulmonary vascular lesions). Here we ask the question: which forms of pulmonary vascular remodeling are reversible and can such remodeling caused by angiogenic proliferation of EC be reversed? It is important to emphasize that the report showing reduction of vascular remodeling by PDGF receptor tyrosine kinase inhibitor showed only a reduction of the pulmonary artery muscularization in chronic hypoxia and monocrotaline models, which lack the feature of clustered proliferated EC in the lumen of pulmonary arteries. The regression of vascular muscularization is an important manifestation, whereby proliferative adult SMC convert back to a nonproliferative state. In contrast, in vitro experiments assessing the contribution of EC to the development of PAH demonstrated that phenotypically altered EC generated as a consequence of a vascular endothelial growth factor receptor blockade did not reverse to normal EC. Whereas it is suggested that the proliferative state of SMC may be reversible, it remains unknown whether phenotypically altered EC can switch back to a normal monolayer-forming EC. This article reviews the pathogenetic concepts of severe PAH and explains the many forms in PAH with reversible or irreversible remodeling.
Collapse
Affiliation(s)
- Seiichiro Sakao
- Department of Respirology (B2), Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, Japan.
| | | | | |
Collapse
|
26
|
Firth AL, Yao W, Remillard CV, Ogawa A, Yuan JXJ. Upregulation of Oct-4 isoforms in pulmonary artery smooth muscle cells from patients with pulmonary arterial hypertension. Am J Physiol Lung Cell Mol Physiol 2010; 298:L548-57. [PMID: 20139178 DOI: 10.1152/ajplung.00314.2009] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Oct-4 is a transcription factor considered to be one of the defining pluripotency markers in embryonic stem cells. Its expression has also been demonstrated in adult stem cells, tumorigenic cells, and, most recently and controversially, in somatic cells. Oct-4 pseudogenes also contribute to carcinogenesis. Oct-4 may be involved in the excessive proliferation of pulmonary arterial smooth muscle cells (PASMC) in patients with idiopathic pulmonary arterial hypertension (IPAH), contributing to the pathogenesis of IPAH. In this study, we show that Oct-4 isoforms are upregulated in IPAH-PASMC. Human embryonic stem cells (H9 line) and human PASMC from normotensive subjects were used throughout the investigation as positive and negative controls. In addition to significant upregulation of Oct-4 in a population of IPAH-PASMC, HIF-2alpha, a hypoxia-inducible transcription factor that has been shown to bind to the Oct-4 promoter and induces its expression and transcriptional activity, was also increased. Interestingly, a substantial upregulation of Oct-4 isoforms and HIF-2alpha was also observed in normal PASMC exposed to chronic hypoxia. In conclusion, the data suggest that both Oct-4 isoforms are upregulated and potentially have a significant role in the development of vascular abnormalities associated with the pathogenesis of IPAH and in pulmonary hypertension triggered by chronic hypoxia.
Collapse
Affiliation(s)
- Amy L Firth
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, California 92093-0725, USA
| | | | | | | | | |
Collapse
|
27
|
Seidel P, Merfort I, Hughes JM, Oliver BGG, Tamm M, Roth M. Dimethylfumarate inhibits NF-{kappa}B function at multiple levels to limit airway smooth muscle cell cytokine secretion. Am J Physiol Lung Cell Mol Physiol 2009; 297:L326-39. [PMID: 19465513 DOI: 10.1152/ajplung.90624.2008] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The antipsoriatic dimethylfumarate (DMF) has been anecdotically reported to reduce asthma symptoms and to improve quality of life of asthma patients. DMF decreases the expression of proinflammatory mediators by inhibiting the transcription factor NF-kappaB and might therefore be of interest for the therapy of inflammatory lung diseases. In this study, we determined the effect of DMF on platelet-derived growth factor (PDGF)-BB- and TNFalpha-induced asthma-relevant cytokines and NF-kappaB activation by primary human asthmatic and nonasthmatic airway smooth muscle cells (ASMC). Confluent nonasthmatic and asthmatic ASMC were incubated with DMF (0.1-100 microM) and/or dexamethasone (0.0001-0.1 microM), NF-kappaB p65 siRNA (100 nM), the NF-kappaB inhibitor helenalin (1 microM) before stimulation with PDGF-BB or TNFalpha (10 ng/ml). Cytokine release was measured by ELISA. NF-kappaB, mitogen and stress-activated kinase (MSK-1), and CREB activation was determined by immunoblotting and EMSA. TNFalpha-induced eotaxin, RANTES, and IL-6 as well as PDGF-BB-induced IL-6 expression was inhibited by DMF and by dexamethasone from asthmatic and nonasthmatic ASMC, but the combination of both drugs showed no glucocorticoid sparing effect in either of the two groups. NF-kappaB p65 siRNA and/or the NF-kappaB inhibitor helenalin reduced PDGF-BB- and TNFalpha-induced cytokine expression, suggesting the involvement of NF-kappaB signaling. DMF inhibited TNFalpha-induced NF-kappaB p65 phosphorylation, NF-kappaB nuclear entry, and NF-kappaB-DNA complex formation, whereas PDGF-BB appeared not to activate NF-kappaB within 60 min. Both stimuli induced the phosphorylation of MSK-1, NF-kappaB p65 at Ser276, and CREB, and all were inhibited by DMF. These data suggest that DMF downregulates cytokine secretion not only by inhibiting NF-kappaB but a wider range of NF-kappaB-linked signaling proteins, which may explain its potential beneficial effect in asthma.
Collapse
Affiliation(s)
- P Seidel
- Department of Research and Pneumology, University Hospital Basel, Switzerland
| | | | | | | | | | | |
Collapse
|
28
|
Fujita Y, Wideman RD, Speck M, Asadi A, King DS, Webber TD, Haneda M, Kieffer TJ. Incretin release from gut is acutely enhanced by sugar but not by sweeteners in vivo. Am J Physiol Endocrinol Metab 2009; 296:E473-9. [PMID: 19106249 DOI: 10.1152/ajpendo.90636.2008] [Citation(s) in RCA: 147] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-1 (GLP-1) are released during meals from endocrine cells located in the gut mucosa and stimulate insulin secretion from pancreatic beta-cells in a glucose-dependent manner. Although the gut epithelium senses luminal sugars, the mechanism of sugar sensing and its downstream events coupled to the release of the incretin hormones are not clearly elucidated. Recently, it was reported that sucralose, a sweetener that activates the sweet receptors of taste buds, triggers incretin release from a murine enteroendocrine cell line in vitro. We confirmed that immunoreactivity of alpha-gustducin, a key G-coupled protein involved in taste sensing, is sometimes colocalized with GIP in rat duodenum. We investigated whether secretion of incretins in response to carbohydrates is mediated via taste receptors by feeding rats the sweet-tasting compounds saccharin, acesulfame potassium, d-tryptophan, sucralose, or stevia. Oral gavage of these sweeteners did not reduce the blood glucose excursion to a subsequent intraperitoneal glucose tolerance test. Neither oral sucralose nor oral stevia reduced blood glucose levels in Zucker diabetic fatty rats. Finally, whereas oral glucose increased plasma GIP levels approximately 4-fold and GLP-1 levels approximately 2.5-fold postadministration, none of the sweeteners tested significantly increased levels of these incretins. Collectively, our findings do not support the concept that release of incretins from enteroendocrine cells is triggered by carbohydrates via a pathway identical to the sensation of "sweet taste" in the tongue.
Collapse
Affiliation(s)
- Yukihiro Fujita
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada V6T 1Z3
| | | | | | | | | | | | | | | |
Collapse
|