Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Eur J Pharmacol ; 973: 176564, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38614383

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 ß and its signaling pathways, STAT3 and nuclear factor-κB (NF-κB). In addition, CRA attenuated the phosphorylation of PDGF receptor ß and STAT3 following the PDGF-BB simulation. The expression and phosphorylation levels of PDGF receptor ß 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 ß-STAT3 and PDGF-PDGF receptor ß-NF-κB signaling axis in macrophages and PASMCs, and thus, ameliorated pulmonary vascular remodeling in PAH.


Subject(s)
Cell Movement , Cell Proliferation , Macrophages , Myocytes, Smooth Muscle , STAT3 Transcription Factor , Signal Transduction , Triterpenes , Triterpenes/pharmacology , Triterpenes/therapeutic use , Animals , Signal Transduction/drug effects , Humans , STAT3 Transcription Factor/metabolism , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Rats , Macrophages/drug effects , Macrophages/metabolism , Male , Cell Movement/drug effects , Cell Proliferation/drug effects , Rats, Sprague-Dawley , Pulmonary Artery/drug effects , Pulmonary Artery/pathology , Pulmonary Artery/metabolism , Platelet-Derived Growth Factor/metabolism , Cell Survival/drug effects , Monocrotaline , Pulmonary Arterial Hypertension/drug therapy , Pulmonary Arterial Hypertension/metabolism , Pulmonary Arterial Hypertension/pathology , Becaplermin/pharmacology , Vascular Remodeling/drug effects , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/pathology
2.
Front Cardiovasc Med ; 11: 1343804, 2024.
Article in English | MEDLINE | ID: mdl-38410243

ABSTRACT

Background: Pulmonary arterial hypertension (PAH) is a severe and rare disease in the cardiopulmonary system. Its pathogenesis involves vascular remodeling of the pulmonary artery, which results in progressive increases in pulmonary arterial pressure. Chronically increased pulmonary arterial pressure causes right ventricular hypertrophy and subsequent right heart failure. Pulmonary vascular remodeling is attributed to the excessive proliferation and migration of pulmonary arterial smooth muscle cells (PASMCs), which are induced by enhanced Ca2+ signaling following the up-/down-regulation of ion channel expression. Objectives: In the present study, the functional expression of two-pore domain potassium KCNK channels was investigated in PASMCs from idiopathic PAH (IPAH) patients and experimental pulmonary hypertensive (PH) animals. Results: In IPAH-PASMCs, the expression of KCNK1/TWIK1 and KCNK2/TREK1 channels was up-regulated, whereas that of KCNK3/TASK1 and KCNK6/TWIK2 channels was down-regulated. The similar up-regulated expression of KCNK1 and KCNK2 channels was observed in the pulmonary arterial smooth muscles of monocrotaline-induced PH rats, Sugen 5416/hypoxia-induced PH rats, and hypoxia-induced PH mice. The facilitated proliferation of IPAH-PASMCs was suppressed by the KCNK channel blockers, quinine and tetrapentylammonium. The migration of IPAH-PASMCs was also suppressed by these channel blockers. Furthermore, increases in the proliferation and migration were inhibited by the siRNA knockdown of KCNK1 or KCNK2 channels. The siRNA knockdown also caused membrane depolarization and subsequent decrease in cytosolic [Ca2+]. The phosphorylated level of c-Jun N-terminal kinase (JNK) was elevated in IPAH-PASMCs compared to normal-PASMCs. The increased phosphorylation was significantly reduced by the siRNA knockdown of KCNK1 or KCNK2 channels. Conclusion: Collectively, these findings indicate that the up-regulated expression of KCNK1 and KCNK2 channels facilitates the proliferation and migration of PASMCs via enhanced Ca2+ signaling and JNK signaling pathway, which is associated with vascular remodeling in PAH.

3.
J Pharmacol Sci ; 153(2): 84-88, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37640473

ABSTRACT

Pulmonary vessels play a pivotal role in oxygen circulation. We previously demonstrated that pimaric acid (PiMA) activated large-conductance Ca2+-activated K+ (BKCa) channels and inhibited voltage-dependent Ca2+ channels (VDCCs). In the present study, PiMA attenuated vasoconstriction induced by high K+ or endothelin-1 in rat pulmonary arterial smooth muscles (PASMs). PiMA also reduced high K+-induced cytosolic [Ca2+] increase in PASM cells. PiMA increased BKCa currents and decreased VDCC currents. BKCa channels and VDCCs were formed by the α/ß1 and α1C/α1D/ß2/ß3 subunits, respectively. These results indicate that PiMA induces vasorelaxation through the dual effects of BKCa channel activation and VDCC inhibition in PASMs.


Subject(s)
Hypertension, Pulmonary , Vasoconstriction , Animals , Rats , Calcium Channels, L-Type , Potassium Iodide , Muscle, Smooth
4.
J Pharmacol Sci ; 151(2): 119-127, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36707177

ABSTRACT

Pulmonary arterial hypertension (PAH) is a progressive and fatal disease that is characterized by vascular remodeling of the pulmonary artery. PAH remodeling is primarily caused by the excessive proliferation of pulmonary arterial smooth muscle cells (PASMCs). Therefore, an inhibitory mechanism is expected as a target for the treatment of PAH. Corosolic acid (CRA) is a pentacyclic triterpenoid extracted from the leaves of Banaba (Lagerstroemia speciosa) that exerts anti-diabetic, anti-inflammatory, and anti-tumor effects. In the present study, the effects of CRA on PAH remodeling were examined using PASMCs from idiopathic pulmonary arterial hypertension (IPAH) patients and monocrotaline (MCT)-induced pulmonary hypertensive (PH) rats. CRA inhibited the excessive proliferation of IPAH-PASMCs in a concentration-dependent manner (IC50 = 14.1 µM). It also reduced the migration of IPAH-PASMCs. The CRA treatment downregulated the expression of signal transducer and activator of transcription 3 (STAT3) in IPAH-PASMCs. In MCT-PH rats, the administration of CRA (1 mg/kg/day) attenuated increases in right ventricular systolic pressure, pulmonary vascular remodeling, and right ventricular hypertrophy. CRA also decreased the expression of STAT3 in pulmonary arterial smooth muscles from MCT-PH rats. In conclusion, the anti-proliferative and anti-migratory effects of CRA in PASMCs ameliorated PAH remodeling by downregulating STAT3 signaling pathways.


Subject(s)
Hypertension, Pulmonary , Pulmonary Arterial Hypertension , Rats , Animals , Pulmonary Arterial Hypertension/drug therapy , Pulmonary Arterial Hypertension/metabolism , Pulmonary Arterial Hypertension/pathology , Familial Primary Pulmonary Hypertension/metabolism , Familial Primary Pulmonary Hypertension/pathology , Hypertension, Pulmonary/metabolism , Down-Regulation , Vascular Remodeling , STAT3 Transcription Factor/metabolism , Pulmonary Artery , Myocytes, Smooth Muscle , Cell Proliferation
5.
Biol Pharm Bull ; 45(11): 1684-1691, 2022 Nov 01.
Article in English | MEDLINE | ID: mdl-35989293

ABSTRACT

Pulmonary arterial hypertension (PAH) is characterized by vascular remodeling of the pulmonary artery, which is mainly attributed to the excessive proliferation of pulmonary arterial smooth muscle cells (PASMCs) comprising the medial layer of pulmonary arteries. The activity of ion channels associated with cytosolic Ca2+ signaling regulates the pathogenesis of PAH. Limited information is currently available on the role of Cl- channels in PASMCs. Therefore, the functional expression of ClC3 channels/transporters was herein investigated in the PASMCs of normal subjects and patients with idiopathic pulmonary arterial hypertension (IPAH). Expression analyses revealed the upregulated expression of ClC3 channels/transporters at the mRNA and protein levels in IPAH-PASMCs. Hypoosmotic perfusion (230 mOsm) evoked swelling-activated Cl- currents (ICl-swell) in normal-PASMCs, whereas 100 µM 4,4'-diisothiocyanatostilbene-2,2'-disulfonic acid (DIDS) exerted the opposite effects. The small interfering RNA (siRNA) knockdown of ClC3 did not affect ICl-swell. On the other hand, ICl-swell was larger in IPAH-PASMCs and inhibited by DIDS and the siRNA knockdown of ClC3. IPAH-PASMCs grew more than normal-PASMCs. The growth of IPAH-PASMCs was suppressed by niflumic acid and DIDS, but not by 9-anthracenecarboxylic acid or T16Ainh-A01. The siRNA knockdown of ClC3 also inhibited the proliferation of IPAH-PASMCs. Collectively, the present results indicate that upregulated ClC3 channels/transporters are involved in ICl-swell and the excessive proliferation of IPAH-PASMCs, thereby contributing to the pathogenesis of PAH. Therefore, ClC3 channels/transporters have potential as a target of therapeutic drugs for the treatment of PAH.


Subject(s)
Myocytes, Smooth Muscle , Humans , Familial Primary Pulmonary Hypertension/drug therapy , Familial Primary Pulmonary Hypertension/genetics , Familial Primary Pulmonary Hypertension/pathology , 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/pharmacology , RNA, Small Interfering/pharmacology , Myocytes, Smooth Muscle/metabolism , Cell Proliferation , Cells, Cultured
6.
Biochem Biophys Res Commun ; 607: 44-48, 2022 06 04.
Article in English | MEDLINE | ID: mdl-35366542

ABSTRACT

In pulmonary arterial smooth muscle cells (PASMCs), an increase in the cytosolic Ca2+ concentration ([Ca2+]cyt) is involved in many physiological processes such as cell contraction and proliferation. However, chronic [Ca2+]cyt increases cause pulmonary vasoconstriction and vascular remodeling, resulting in pulmonary arterial hypertension (PAH). Therefore, [Ca2+]cyt signaling plays a substantial role in the regulation of physiological and pathological functions in PASMCs. In the present study, the effects of SKF96365 on [Ca2+]cyt were examined in PASMCs from normal subjects and idiopathic pulmonary arterial hypertension (IPAH) patients. SKF96365 is widely used as a blocker of non-selective cation channels. SKF96365 did not affect the resting [Ca2+]cyt in normal-PASMCs. However, SKF96365 increased [Ca2+]cyt in IPAH-PASMCs in a concentration-dependent manner (EC50 = 18 µM). The expression of Ca2+-sensing receptors (CaSRs) was higher in IPAH-PASMCs than in normal-PASMCs. The SKF96365-induced [Ca2+]cyt increase was inhibited by CaSR antagonists, NPS2143 and Calhex 231. The CaSR-mediated [Ca2+]cyt increase was facilitated by SKF96365 and the activation was blocked by NPS2143 or Calhex 231. In addition, the SKF96365-induced [Ca2+]cyt increase was reduced by siRNA knockdown of CaSRs. Taken together, SKF96365 activates CaSRs in IPAH-PASMCs and promotes [Ca2+]cyt signaling.


Subject(s)
Hypertension, Pulmonary , Receptors, Calcium-Sensing , Calcium/metabolism , Cell Proliferation , Cells, Cultured , Familial Primary Pulmonary Hypertension/pathology , Humans , Hypertension, Pulmonary/metabolism , Imidazoles , Myocytes, Smooth Muscle/metabolism , Pulmonary Artery/pathology , Receptors, Calcium-Sensing/metabolism
7.
Life Sci ; 288: 120171, 2022 Jan 01.
Article in English | MEDLINE | ID: mdl-34822800

ABSTRACT

AIM: The abnormal expression of oncogenic tyrosine kinase receptors such as platelet-derived growth factor receptors (PDGFRs) has been reported in cancer progression. However, the role of PDGFRs in the human androgen-independent prostate cancer PC-3 cell line is not well understood. Thus, this study examined the role of PDGFRs in androgen-independent PC-3 cells. MAIN METHODS: PDGFR mRNA and protein expression was determined by quantitative real-time PCR and western blotting, respectively. The effects of the tyrosine kinase inhibitor imatinib (imatinib mesylate) and small interfering RNAs (siRNAs) were determined by a Cell Counting Kit-8 assay, bromodeoxyuridine assay, and Transwell migration assay. The in vivo effect of imatinib was analyzed using a tumor formation assay in nude mice. KEY FINDINGS: PDGFRα was upregulated in androgen-independent PC-3 cells compared with normal prostate epithelial cells. PDGF-BB induced the phosphorylation of PDGFRα and downstream signaling molecules, including Akt, in a dose-dependent manner. Imatinib reduced the phosphorylation of the PDGFRα/Akt axis. Imatinib also suppressed the viability, proliferation, migration, and tumor growth of PC-3 cells. PDGFRα knockdown by siRNA decreased the viability and migration of PC-3 cells. SIGNIFICANCE: These results demonstrated the distinct contribution of PDGFRα signaling to the proliferation and migration of PC-3 cells and suggested the potential for PDGFRα as a therapeutic target for metastatic and androgen-independent prostate cancer.


Subject(s)
Cell Movement , Cell Proliferation , Gene Expression Regulation, Neoplastic/drug effects , Imatinib Mesylate/pharmacology , Prostatic Neoplasms/prevention & control , RNA, Small Interfering/genetics , Receptor, Platelet-Derived Growth Factor alpha/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , PC-3 Cells , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Receptor, Platelet-Derived Growth Factor alpha/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
8.
J Pharmacol Sci ; 147(1): 81-85, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34294376

ABSTRACT

Pulmonary arterial hypertension (PAH) is a rare, progressive, and fatal cardiovascular/lung disease. The incidence rate is affected by age. Monocrotaline (MCT, 60 mg/kg)-treated rats are widely used as an experimental PAH model. Here, we found that young rats died at a mean of 23.4 days after MCT injection, whereas adult rats survived for over 42 days. However, young (7-week-old) and adult (20-week-old) MCT-treated rats developed PAH, and had upregulated Ca2+-sensing receptor and transient receptor potential canonical subfamily 6 channel expression in pulmonary arteries. The present study provides novel information for elucidating the mechanism underlying the age difference in PAH patients.


Subject(s)
Hypertension, Pulmonary/metabolism , Monocrotaline/adverse effects , Adult , Age Factors , Animals , Calcium Channels/metabolism , Disease Models, Animal , Female , Humans , Hypertension, Pulmonary/chemically induced , Male , Middle Aged , Pulmonary Artery/metabolism , Rats, Sprague-Dawley , Receptors, Calcium-Sensing/metabolism
9.
Nihon Yakurigaku Zasshi ; 156(3): 145, 2021.
Article in Japanese | MEDLINE | ID: mdl-33952841
10.
Nihon Yakurigaku Zasshi ; 156(3): 161-165, 2021.
Article in Japanese | MEDLINE | ID: mdl-33952845

ABSTRACT

Pulmonary hypertension (PH) is defined as mean pulmonary arterial pressure at rest ≥25 mmHg. Pulmonary arterial hypertension (PAH) is classified as group 1 of PH and is a progressive and fatal disease of the pulmonary artery. The pathogenesis is sustained pulmonary vasoconstriction and pulmonary vascular remodeling, which cause progressive elevations in pulmonary vascular resistance and pulmonary arterial pressure. Elevated pulmonary arterial pressure leads to right heart failure and finally death. The pulmonary vascular remodeling is triggered by an increase in cytosolic Ca2+ concentration ([Ca2+]cyt). [Ca2+]cyt is regulated by the stimulation of vasoconstrictors and growth factors though their receptors and ion channels on the plasma membrane. It has been reported that the epidermal growth factor (EGF), fibroblast growth factor (FGF), insulin-like growth factor (IGF), vascular endothelial growth factor (VEGF), and platelet-derived growth factor (PDGF) are involved in the development of PAH. Upon binding of these growth factors with their specific receptor tyrosine kinases, their receptors activate cytosolic Ca2+ signaling and signal transduction cascades to induce cell proliferation, differentiation, and migration. Expressions of some growth factors and their receptors upregulate in PAH patients, which contributes to the formation of vascular remodeling and plexiform lesions in PAH. We have recently found that enhanced Ca2+-sensing receptor (CaSR) function is involved the development of PAH and CaSR expression is upregulated by PDGF in pulmonary arterial smooth muscle cells (PASMCs) from idiopathic PAH patients. This review will be discussed the physiological and pathological roles of growth factors in PAH.


Subject(s)
Hypertension, Pulmonary , Vascular Remodeling , Calcium , Cell Proliferation , Humans , Muscle, Smooth, Vascular , Pulmonary Artery , Vascular Endothelial Growth Factor A
11.
Front Pharmacol ; 12: 667474, 2021.
Article in English | MEDLINE | ID: mdl-33959030

ABSTRACT

Vascular endothelial growth factor (VEGF) signaling plays a critical role in the carcinogenesis and tumor development of several cancer types. However, its pathological significance in prostate cancer, one of the most frequent and lethal malignancies in men, remains unclear. In the present study, we focused on a pathological role of the VEGF receptors (VEGFRs), and examined their expression and effects of MAZ51 (an inhibitor of the tyrosine kinase of VEGFR-3) on cell proliferation, migration, and tumor growth in human prostate cancer cells. The expression level of VEGFR-3 was higher in androgen-independent and highly metastatic prostate cancer PC-3 cells than in other prostate PrEC, LNCaP, and DU145 cells. In PC-3 cells, VEGFR-3 and Akt were phosphorylated following a stimulation with 50 ng/ml VEGF-C, and these phosphorylations were blocked by 3 µM MAZ51. Interestingly, PC-3 cells themselves secreted VEGF-C, which was markedly larger amount compared with PrEC, LNCaP, and DU145 cells. MAZ51 reduced the expression of VEGFR-3 but not VEGFR-1 and VEGFR-2. The proliferation of PC-3 cells was inhibited by MAZ51 (IC50 = 2.7 µM) and VEGFR-3 siRNA, and partly decreased by 100 nM GSK690693 (an Akt inhibitor) and 300 nM VEGFR2 Kinase Inhibitor I. MAZ51 and VEGFR-3 siRNA also attenuated the VEGF-C-induced migration of PC-3 cells. Moreover, MAZ51 blocked the tumor growth of PC-3 cells in a xenograft mouse model. These results suggest that VEGFR-3 signaling contributes to the cell proliferation, migration, and tumor growth of androgen-independent/highly metastatic prostate cancer. Therefore, the inhibition of VEGFR-3 has potential as a novel therapeutic target for the treatment for prostate cancer.

12.
Transl Res ; 233: 127-143, 2021 07.
Article in English | MEDLINE | ID: mdl-33691194

ABSTRACT

Ophiocordyceps sinensis (OCS), an entomopathogenic fungus, is known to exert antiproliferative and antitissue remodeling effects. Vascular remodeling and vasoconstriction play critical roles in the development of pulmonary hypertension (PH). The therapeutic potential of OCS for PH was investigated using rodent PH models, and cultured pulmonary artery endothelial and smooth muscle cells (PAECs and PASMCs), with a focus on the involvement of TRPM7. OCS ameliorated the development of PH, right ventricular hypertrophy and dysfunction in the monocrotaline-induced PH rats. The genetic knockout of TRPM7 attenuated the development of PH in mice with monocrotaline pyrrole-induced PH. TRPM7 was associated with medial hypertrophy and the plexiform lesions in rats and humans with PH. OCS suppressed proliferation of PASMCs derived from the PH patients. Ethanol extracts of OCS inhibited TRPM7-like current, TGF-ß2-induced endothelial-mesenchymal transition, IL-6-induced STAT3 phosphorylation, and PDGF-induced Akt phosphorylation in PAECs or PASMCs. These inhibitory effects were recapitulated by either siRNA-mediated TRPM7 knockdown or treatment with TRPM7 antagonist FTY-720. OCS and FTY-720 induced vasorelaxation in the isolated normal human pulmonary artery. As a result, the present study proposes the therapeutic potential of OCS for the treatment of PH. The inhibition of TRPM7 is suggested to underlie the therapeutic effect of OCS.


Subject(s)
Cordyceps/physiology , Hypertension, Pulmonary/physiopathology , Hypertension, Pulmonary/therapy , TRPM Cation Channels/antagonists & inhibitors , Animals , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Fingolimod Hydrochloride/pharmacology , Gene Knockdown Techniques , Humans , Hypertension, Pulmonary/pathology , Male , Medicine, Chinese Traditional , Mice , Mice, Knockout , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/physiology , Proto-Oncogene Proteins c-akt/metabolism , Pulmonary Artery/pathology , Pulmonary Artery/physiopathology , Rats , Rats, Sprague-Dawley , STAT3 Transcription Factor/metabolism , TRPM Cation Channels/deficiency , TRPM Cation Channels/genetics , TRPM Cation Channels/physiology , Translational Research, Biomedical , Vasodilation
13.
Biochem Biophys Res Commun ; 534: 795-801, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33160621

ABSTRACT

Pulmonary arterial hypertension (PAH) is a progressive and fatal disease that is characterized by the irreversible remodeling of the pulmonary artery. Although several PAH drugs have been developed, additional drugs are needed. Rho kinases (ROCKs) are involved in the pathogenesis of PAH, and thus, their inhibitors may prevent the development of PAH. However, the therapeutic benefits of ROCK isoform-specific inhibitors for PAH remain largely unknown. The in vitro and in vivo effects of the ROCK2-specific inhibitor, KD025, were examined herein using pulmonary arterial smooth muscle cells (PASMCs) from idiopathic pulmonary arterial hypertension (IPAH) patients and monocrotaline (MCT)-induced pulmonary hypertensive (PH) rats. The expression of ROCK1 was similar between normal- and IPAH-PASMCs, whereas that of ROCK2 was markedly higher in IPAH-PASMCs than in normal-PASMCs. KD025 inhibited the accelerated proliferation of IPAH-PASMCs in a concentration-dependent manner (IC50 = 289 nM). Accelerated proliferation was also reduced by the siRNA knockdown of ROCK2. In MCT-PH rats, the expression of ROCK2 was up-regulated in PASMCs. Elevated right ventricular systolic pressure in MCT-PH rats was attenuated by KD025 (1 mg/kg/day). These results strongly suggest that enhanced ROCK2 signaling is involved in the pathogenic mechanism underlying the development of PAH, including accelerated PASMC proliferation and vascular remodeling in patients with PAH. Therefore, ROCK2 may be a novel therapeutic target for the treatment of PAH.


Subject(s)
Familial Primary Pulmonary Hypertension/pathology , Heterocyclic Compounds, 4 or More Rings/pharmacology , Pulmonary Arterial Hypertension/drug therapy , rho-Associated Kinases/genetics , Animals , Cell Line , Cell Proliferation/drug effects , Familial Primary Pulmonary Hypertension/enzymology , Humans , Male , Monocrotaline/toxicity , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Pulmonary Arterial Hypertension/chemically induced , Pulmonary Arterial Hypertension/physiopathology , Pulmonary Artery/physiopathology , Rats, Sprague-Dawley , Up-Regulation , Vascular Remodeling , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/metabolism
14.
J Mol Cell Cardiol ; 148: 50-62, 2020 11.
Article in English | MEDLINE | ID: mdl-32889002

ABSTRACT

Pulmonary arterial hypertension (PAH) is a multifactorial disease characterized by pulmonary arterial vasoconstriction and remodeling. Src family tyrosine kinases, including Fyn, play critical roles in vascular remodeling via the inhibition of STAT3 signaling. EPA is known to inhibit Fyn kinase activity. This study investigated the therapeutic potential and underlying mechanisms of EPA and its metabolite, resolvin E1 (RvE1), to treat PAH using monocrotaline-induced PAH model rats (MCT-PAH), human pulmonary artery endothelial cells (HPAECs), and human pulmonary artery smooth muscle cells (HPASMCs). Administration of EPA 1 and 2 weeks after MCT injection both ameliorated right ventricular hypertrophy, remodeling and dysfunction, and medial wall thickening of the pulmonary arteries and prolonged survival in MCT-PAH rats. EPA attenuated the enhanced contractile response to 5-hydroxytryptamine in isolated pulmonary arteries of MCT-PAH rats. Mechanistically, the treatment with EPA and RvE1 or the introduction of dominant-negative Fyn prevented TGF-ß2-induced endothelial-to-mesenchymal transition and IL-6-induced phosphorylation of STAT3 in cultured HPAECs. EPA and RvE1 suppressed Src family kinases' activity as evaluated by their phosphorylation status in cultured HPAECs and HPASMCs. EPA and RvE1 suppressed vasocontraction of rat and human PA. Furthermore, EPA and RvE1 inhibited the enhanced proliferation and activity of Src family kinases in HPASMCs derived from patients with idiopathic PAH. EPA ameliorated PAH's pathophysiology by mitigating vascular remodeling and vasoconstriction, probably inhibiting Src family kinases, especially Fyn. Thus, EPA is considered a potent therapeutic agent for the treatment of PAH.


Subject(s)
Eicosapentaenoic Acid/therapeutic use , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/enzymology , Proto-Oncogene Proteins c-fyn/antagonists & inhibitors , Animals , Cell Proliferation/drug effects , Eicosapentaenoic Acid/analogs & derivatives , Eicosapentaenoic Acid/pharmacology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Endothelium, Vascular/physiopathology , Humans , Hypertension, Pulmonary/physiopathology , Hypertrophy, Right Ventricular/complications , Hypertrophy, Right Ventricular/physiopathology , Interleukin-6/pharmacology , Male , Mesoderm/drug effects , Mesoderm/pathology , Mesoderm/physiopathology , Monocrotaline , Myocardial Contraction/drug effects , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-fyn/metabolism , Pulmonary Artery/drug effects , Pulmonary Artery/physiopathology , Rats, Sprague-Dawley , STAT3 Transcription Factor/metabolism , Survival Analysis , Transforming Growth Factor beta2/pharmacology , Vasodilation/drug effects , Ventricular Remodeling/drug effects , src-Family Kinases/metabolism
15.
J Physiol Sci ; 70(1): 44, 2020 Sep 22.
Article in English | MEDLINE | ID: mdl-32962633

ABSTRACT

Glucose uptake and adenosine triphosphate (ATP) generation are important for the survival and growth of endothelial cells. An increase of glucose uptake under hypoxia was previously shown to be associated with the increased expression of glucose transporters (GLUTs). However, the regulation of GLUT trafficking to the cell surface has not been examined in detail. Here, we report the characterization of GLUT1 translocation to the plasma membrane during hypoxia in endothelial cells. Human umbilical vein endothelial cells (HUVECs) were exposed to hypoxia (1% O2) for 12 h, which significantly induced GLUT1 expression and translocation to the plasma membrane. GLUT1 translocation was associated with a decrease of intracellular ATP by hypoxia. Decreasing ATP levels with antimycin-A and 2-deoxyglucose induced GLUT1 translocation under normoxia. The induction of hypoxia-inducible factor-1α under normoxia did not influence the cell surface expression of GLUT1 or cellular ATP concentration. Interestingly, the translocation of GLUT1 induced by hypoxia was inhibited by the ATP-sensitive potassium (KATP) channel inhibitor glibenclamide, while the mitochondrial KATP channel inhibitor 5-HD did not influence GLUT1 translocation during hypoxia. These observations indicate that a decrease of intracellular ATP triggers GLUT1 translocation to the plasma membrane and is mediated by KATP channels, which would contribute to glucose uptake in HUVECs during hypoxia.


Subject(s)
Cell Membrane/metabolism , Deoxyglucose/metabolism , Glucose Transporter Type 1/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Adenosine Triphosphate/metabolism , Cell Hypoxia , Cells, Cultured , Glucose Transporter Type 1/genetics , Humans , KATP Channels/genetics , KATP Channels/metabolism , Protein Transport
16.
Nihon Yakurigaku Zasshi ; 155(4): 230-235, 2020.
Article in Japanese | MEDLINE | ID: mdl-32612035

ABSTRACT

Pulmonary arterial hypertension (PAH) is a progressive and lethal disease of the pulmonary artery. The pathogenesis of PAH is mainly sustained vasoconstriction and vascular remodeling of the pulmonary artery. These pathogeneses cause progressive elevations in pulmonary vascular resistance and pulmonary arterial pressure in PAH patients. Elevated pulmonary arterial pressure leads to right heart failure and finally death. The vascular remodeling is caused by the enhanced proliferation and reduced apoptosis of pulmonary arterial smooth muscle cells (PASMCs). Excitable abnormality in the pulmonary artery of PAH patients is mostly mediated by an elevated cytosolic Ca2+ concentration. PASMCs express several Ca2+-permeable channels including voltage-dependent Ca2+ channels, store-operated Ca2+ (SOC) channels, and receptor-operated Ca2+ (ROC) channels. The activation and upregulation of these Ca2+ channels have been reported in PASMCs from PAH patients. Here, we analyzed pathophysiological functions of enhanced Ca2+ signaling mediated by SOC and ROC channels using PASMCs from idiopathic PAH patients and animal PAH models. Notch signal enhanced transient receptor potential canonical 6 (TRPC6) "SOC" channels via direct (non-genomic and stimulatory) and indirect (genomic and upregulating) effects in PAH. On the other hand, the activation of Ca2+-sensing receptors evoked Ca2+ influx through TRPC6 "ROC" channels in PAH. In addition, TRPC6 channel blocker and TRPC6 gene deletion inhibited the development of PAH. Specifically, TRPC6 channels potentially form both ROC and SOC channels in PASMCs, which are involved in the pathophysiological events in PAH. Therefore, targeting TRPC6 channels in PASMCs may help develop novel therapeutic approach for PAH.


Subject(s)
Hypertension, Pulmonary , Pulmonary Arterial Hypertension , Transient Receptor Potential Channels , Animals , Calcium/metabolism , Cell Proliferation , Humans , Muscle, Smooth, Vascular/metabolism , Pulmonary Artery/metabolism , TRPC6 Cation Channel/genetics
17.
Am J Physiol Lung Cell Mol Physiol ; 318(1): L10-L26, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31553627

ABSTRACT

Downregulated expression of K+ channels and decreased K+ currents in pulmonary artery smooth muscle cells (PASMC) have been implicated in the development of sustained pulmonary vasoconstriction and vascular remodeling in patients with idiopathic pulmonary arterial hypertension (IPAH). However, it is unclear exactly how K+ channels are downregulated in IPAH-PASMC. MicroRNAs (miRNAs) are small non-coding RNAs that are capable of posttranscriptionally regulating gene expression by binding to the 3'-untranslated regions of their targeted mRNAs. Here, we report that specific miRNAs are responsible for the decreased K+ channel expression and function in IPAH-PASMC. We identified 3 miRNAs (miR-29b, miR-138, and miR-222) that were highly expressed in IPAH-PASMC in comparison to normal PASMC (>2.5-fold difference). Selectively upregulated miRNAs are correlated with the decreased expression and attenuated activity of K+ channels. Overexpression of miR-29b, miR-138, or miR-222 in normal PASMC significantly decreased whole cell K+ currents and downregulated voltage-gated K+ channel 1.5 (KV1.5/KCNA5) in normal PASMC. Inhibition of miR-29b in IPAH-PASMC completely recovered K+ channel function and KV1.5 expression, while miR-138 and miR-222 had a partial or no effect. Luciferase assays further revealed that KV1.5 is a direct target of miR-29b. Additionally, overexpression of miR-29b in normal PASMC decreased large-conductance Ca2+-activated K+ (BKCa) channel currents and downregulated BKCa channel ß1 subunit (BKCaß1 or KCNMB1) expression, while inhibition of miR-29b in IPAH-PASMC increased BKCa channel activity and BKCaß1 levels. These data indicate upregulated miR-29b contributes at least partially to the attenuated function and expression of KV and BKCa channels in PASMC from patients with IPAH.


Subject(s)
Down-Regulation/genetics , Familial Primary Pulmonary Hypertension/genetics , MicroRNAs/genetics , Potassium Channels, Voltage-Gated/genetics , Adolescent , Adult , Cells, Cultured , Familial Primary Pulmonary Hypertension/metabolism , Female , Humans , Male , Membrane Potentials/genetics , Middle Aged , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Pulmonary Artery/metabolism , RNA, Messenger/genetics , Up-Regulation/genetics , Vasoconstriction/genetics , Young Adult
18.
Front Mol Neurosci ; 12: 175, 2019.
Article in English | MEDLINE | ID: mdl-31379498

ABSTRACT

Maintaining Ca2+ homeostasis in lymphatic fluids is necessary for proper hearing. Despite its significance, the mechanisms that maintain the cochlear lymphatic Ca2+ concentrations within a certain range are not fully clarified. We investigated the functional expression of calcium-sensing receptor (CaSR), which plays a pivotal role in sensing extracellular Ca2+ concentrations for feedback regulations. Western blotting for CaSR revealed an approximately 130-kDa protein expression in cochlear tissue extracts and immunohistochemical analysis revealed its expression specifically in type I fibrocytes in the spiral ligament, fibrocytes in the supralimbal and limbal regions, the epithelium of the osseous spiral lamina, and the smooth muscle cells of the spiral modiolar arteries. Ca2+ imaging demonstrated that extracellular Ca2+ increased the levels of intracellular Ca2+ in CaSR-expressing fibrocytes in the spiral ligament, and that this was suppressed by the CaSR inhibitor, NPS2143. Furthermore, hearing thresholds were moderately elevated by intracochlear application of the CaSR inhibitors NPS2143 and Calhex231, across a range of frequencies (8-32 kHz). These results demonstrate the functional expression of CaSR in the cochlear perilymphatic compartment. In addition, the elevated hearing thresholds that are achieved by inhibiting CaSR suggest this is a required mechanism for normal hearing, presumably by sensing perilymphatic Ca2+ to stabilize Ca2+ concentrations within a certain range. These results provide novel insight into the mechanisms regulating Ca2+ homeostasis in the cochlea and provide a new perspective on cochlear physiology.

19.
FASEB J ; 33(6): 7363-7374, 2019 06.
Article in English | MEDLINE | ID: mdl-30865840

ABSTRACT

Pulmonary arterial hypertension (PAH) is a progressive and fatal disease associated with remodeling of the pulmonary artery. We previously reported that the Ca2+-sensing receptor (CaSR) is up-regulated in pulmonary arterial smooth muscle cells (PASMCs) from patients with idiopathic PAH (IPAH) and contributes to enhanced Ca2+ responses and excessive cell proliferation. However, the mechanisms underlying the up-regulation of CaSR have not yet been elucidated. We herein examined involvement of platelet-derived growth factor (PDGF) on CaSR expression, Ca2+ responses, and proliferation in PASMCs. The expression of PDGF receptors was higher in PASMCs from patients with IPAH than in PASMCs from normal subjects. In addition, PDGF-induced activation of PDGF receptors and their downstream molecules [ERK1/2, p38, protein kinase B, and signal transducer and activator of transcription (STAT) 1/3] were sustained longer in PASMCs from patients with IPAH. The PDGF-induced CaSR up-regulation was attenuated by small interfering RNA knockdown of PDGF receptors and STAT1/3, and by the treatment with imatinib. In monocrotaline-induced pulmonary hypertensive rats, the up-regulation of CaSR was reduced by imatinib. The combination of NPS2143 and imatinib additively inhibited the development of pulmonary hypertension. These results suggest that enhanced PDGF signaling is involved in CaSR up-regulation, leading to excessive PASMC proliferation and vascular remodeling in patients with IPAH. The linkage between CaSR and PDGF signals is a novel pathophysiological mechanism contributing to the development of PAH.-Yamamura, A., Nayeem, M. J., Al Mamun, A., Takahashi, R., Hayashi, H., Sato, M. Platelet-derived growth factor up-regulates Ca2+-sensing receptors in idiopathic pulmonary arterial hypertension.


Subject(s)
Gene Expression Regulation/physiology , Hypertension, Pulmonary/physiopathology , Myocytes, Smooth Muscle/metabolism , Platelet-Derived Growth Factor/physiology , Receptors, Calcium-Sensing/biosynthesis , Vascular Remodeling/physiology , Animals , Calcium/physiology , Cell Division/drug effects , Cells, Cultured , Disease Models, Animal , Gene Expression Regulation/drug effects , Humans , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/pathology , Hypertension, Pulmonary/prevention & control , Imatinib Mesylate/pharmacology , Imatinib Mesylate/therapeutic use , Male , Monocrotaline/toxicity , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/drug effects , Naphthalenes/pharmacology , Naphthalenes/therapeutic use , Platelet-Derived Growth Factor/pharmacology , Pulmonary Artery/pathology , Rats , Rats, Sprague-Dawley , Receptors, Calcium-Sensing/genetics , Receptors, Platelet-Derived Growth Factor/agonists , Receptors, Platelet-Derived Growth Factor/physiology , Signal Transduction/drug effects , Vascular Remodeling/drug effects
20.
Sci Rep ; 9(1): 1560, 2019 02 07.
Article in English | MEDLINE | ID: mdl-30733465

ABSTRACT

Choroidal neovascularization (CNV) is associated with age-related macular degeneration (AMD), a major cause of vision loss among elderly people. Vascular endothelial cell growth factor (VEGF) is essential for the development and progression of AMD, and VEGF signaling molecules are effective targets for the treatment of AMD. We recently reported that activator of G-protein signaling 8 (AGS8), a receptor-independent Gßγ regulator, is involved in VEGF-induced angiogenesis in cultured endothelial cells (EC); however, the role of AGS8 in CNV is not yet understood. This study aimed to explore the role of AGS8 in CNV in cultured cells, explanted choroid tissue, and laser-induced CNV in a mouse AMD model. AGS8 knockdown in cultured choroidal EC inhibited VEGF-induced VEGFR-2 phosphorylation, cell proliferation, and migration. AGS8 knockdown also downregulated cell sprouting from mouse choroidal tissue in ex vivo culture. A mouse model of laser-induced CNV, created to analyze the roles of AGS8 in vivo, demonstrated that AGS8 mRNA was significantly upregulated in choroidal lesions and AGS8 was specifically expressed in the neovasculature. Local AGS8 knockdown in intravitreal tissue significantly inhibited laser-induced AGS8 upregulation and suppressed CNV, suggesting that AGS8 knockdown in the choroid has therapeutic potential for AMD. Together, these results demonstrate that AGS8 plays critical roles in VEGF-induced CNV.


Subject(s)
Choroidal Neovascularization/etiology , Choroidal Neovascularization/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Vascular Endothelial Growth Factor A/metabolism , Animals , Cell Line , Cells, Cultured , Choroidal Neovascularization/pathology , Disease Models, Animal , Disease Susceptibility , Endothelial Cells/metabolism , Gene Expression , Intracellular Signaling Peptides and Proteins/metabolism , Macaca mulatta , Macular Degeneration/etiology , Macular Degeneration/metabolism , Macular Degeneration/pathology , Male , Mice , RNA, Small Interfering/genetics , Vascular Endothelial Growth Factor A/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...