1
|
Valuparampil Varghese M, James J, Bharti D, Rischard F, Rafikova O, Rafikov R. Circulating free heme induces cytokine storm and pulmonary hypertension through the MKK3/p38 axis. Am J Physiol Lung Cell Mol Physiol 2024; 327:L574-L586. [PMID: 39197168 PMCID: PMC11482467 DOI: 10.1152/ajplung.00422.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 08/21/2024] [Accepted: 08/27/2024] [Indexed: 08/30/2024] Open
Abstract
Hemolysis is associated with pulmonary hypertension (PH), but the direct contribution of circulating free heme to the PH pathogenesis remains unclear. Here, we show that the elevated levels of circulating free heme are sufficient to induce PH and inflammatory response in mice and confirm the critical role of mitogen-activated protein kinase kinase-3 (MKK3)-mediated pathway in free heme signaling. Following the continuous infusion of heme for 2 wk, wild-type (WT) but not MKK3 knockout (KO) mice develop PH, as evidenced by a significantly elevated right ventricular (RV) systolic pressure, RV hypertrophy, and pulmonary vascular remodeling. The MKK3/p38 axis, markedly activated by heme infusion in WTs, results in upregulated proliferative/cytokine signaling targets Akt, ERK1/2, and STAT3, which were abrogated in MKK3 KO mice. Moreover, the MKK3 KOs were protected against heme-mediated endothelial barrier dysfunction by restoring the tight junction protein zonula occludens-1 expression and diminishing the inflammatory cell infiltration in the lungs. Plasma cytokine multiplex analysis revealed a severe cytokine storm already 24 h after initiation of heme infusion, with a significant increase of 19 cytokines, including IL-1b, IL-2, IL-6, IL-9, and TNF-a, in WT animals and complete attenuation of cytokine production in MKK3 KO mice. Together, these findings reveal a causative role of circulating free heme in PH through activating inflammatory and proliferative responses. The central role of MKK3 in orchestrating the heme-mediated pathogenic response supports MKK3 as an attractive therapeutic target for PH and other lung inflammatory diseases linked to hemolytic anemia.NEW & NOTEWORTHY This study demonstrates that elevated levels of circulating free heme can induce pulmonary hypertension (PH) and inflammation in mice. Continuous heme infusion activated the MKK3/p38 pathway, leading to increased right ventricular pressure, right ventricular hypertrophy, and vascular remodeling. This activation upregulated signaling cascades such as Akt, ERK1/2, and STAT3, whereas MKK3 knockout mice were protected against these changes and had reduced inflammatory responses, highlighting MKK3's potential as a therapeutic target for PH.
Collapse
Affiliation(s)
- Mathews Valuparampil Varghese
- Division of Pulmonary, Critical Care, Sleep, and Occupational Medicine, Department of Medicine, Indiana University, Indianapolis, Indiana, United States
| | - Joel James
- Division of Pulmonary, Critical Care, Sleep, and Occupational Medicine, Department of Medicine, Indiana University, Indianapolis, Indiana, United States
| | - Dinesh Bharti
- Division of Pulmonary, Critical Care, Sleep, and Occupational Medicine, Department of Medicine, Indiana University, Indianapolis, Indiana, United States
| | - Franz Rischard
- Department of Medicine, The University of Arizona College of Medicine - Tucson, Tucson, Arizona, United States
| | - Olga Rafikova
- Division of Pulmonary, Critical Care, Sleep, and Occupational Medicine, Department of Medicine, Indiana University, Indianapolis, Indiana, United States
| | - Ruslan Rafikov
- Division of Pulmonary, Critical Care, Sleep, and Occupational Medicine, Department of Medicine, Indiana University, Indianapolis, Indiana, United States
| |
Collapse
|
2
|
Encinas-Basurto D, Acosta MF, Eedara BB, Fineman JR, Black SM, Mansour HM. Design and comprehensive characterization of dry powder inhalation aerosols of simvastatin DPPC/DPPG lung surfactant-mimic nanoparticles/microparticles for pulmonary nanomedicine. RSC Adv 2024; 14:29413-29427. [PMID: 39285876 PMCID: PMC11404307 DOI: 10.1039/d4ra04947k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 09/05/2024] [Indexed: 09/19/2024] Open
Abstract
The Rho Kinase (ROCK) pathway is recognized to be involved in changes that lead to remodeling in pulmonary hypertension (PH), particularly cellular processes including signaling, contraction, migration, proliferation, differentiation, and apoptosis. Simvastatin (Sim) has a potent anti-proliferative and pro-apoptotic effect on vasculature smooth muscle cells through the inhibition of the synthesis of isoprenoids intermediates which are essential for the post-translational isoprenylation of Rho, Rac, and Ras family GTPases. Sim targets the underlying mechanism in vascular remodeling. Using bionanomaterials and particle engineering design, this innovative study reports on the advanced inhalable dry powders composed of sim with synthetic phospholipid bionanomaterials, DPPC/DPPG, as a lung surfactant-mimic. These were successfully designed and produced as co-spray dried (Co-SD) nanoparticles and microparticles for nanomedicine delivery as dry powder inhalers (DPIs). Different techniques were used to comprehensively characterize the physicochemical properties of the resulting Co-SD particles. The Next Generation ImpactorTM (NGI™) was used with three different FDA-approved human DPI devices with varying shear stress which were the HandiHaler®, Neohaler®, Aerolizer® DPI devices for aerosol dispersion performance. The formulation-device interactions were examined and correlated. Using human lung cells from different lung regions, in vitro cell viability and transepithelial electrical resistance (TEER) at the air-liquid interface showed biocompatibility of the formulations as a function of dose.
Collapse
Affiliation(s)
- David Encinas-Basurto
- Skaggs Pharmaceutical Sciences Center, College of Pharmacy, The University of Arizona AZ USA
- Biopolymers-CTAOA, Research Center for Food and Development (CIAD, A.C.) Hermosillo Mexico
| | - Maria F Acosta
- Skaggs Pharmaceutical Sciences Center, College of Pharmacy, The University of Arizona AZ USA
| | - Basanth Babu Eedara
- Skaggs Pharmaceutical Sciences Center, College of Pharmacy, The University of Arizona AZ USA
- Center for Translational Science, Florida International University 11350 SW Village Parkway Port St. Lucie FL 34987-2352 USA +7(772) 345-4731
| | - Jeffrey R Fineman
- Department of Pediatrics, University of California San Francisco School of Medicine San Francisco CA USA
| | - Stephen M Black
- Center for Translational Science, Florida International University 11350 SW Village Parkway Port St. Lucie FL 34987-2352 USA +7(772) 345-4731
- Department of Medicine, Division of Translational and Regenerative Medicine, The University of Arizona College of Medicine Tucson AZ USA
| | - Heidi M Mansour
- Skaggs Pharmaceutical Sciences Center, College of Pharmacy, The University of Arizona AZ USA
- Center for Translational Science, Florida International University 11350 SW Village Parkway Port St. Lucie FL 34987-2352 USA +7(772) 345-4731
- Department of Medicine, Division of Translational and Regenerative Medicine, The University of Arizona College of Medicine Tucson AZ USA
- The University of Arizona, BIO5 Institute Tucson AZ USA
| |
Collapse
|
3
|
Xie O, Li SS, Luo Y, Wang L. [Protective effects of 2-methoxyestradiol against hypoxic pulmonary hypertension in neonatal rats]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2024; 26:757-764. [PMID: 39014954 DOI: 10.7499/j.issn.1008-8830.2401078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 07/18/2024]
Abstract
OBJECTIVES To investigate the protective effects of 2-methoxyestradiol (2ME) against hypoxic pulmonary hypertension (HPH) in neonatal rats. METHODS Ninety-six Wistar neonatal rats were randomly divided into a normoxia group, a hypoxia group, and a hypoxia + 2ME group, with each group further subdivided into 3-day, 7-day, 14-day, and 21-day subgroups, containing eight rats each. The hypoxia and hypoxia + 2ME groups received daily subcutaneous injections of saline and 2ME (240 μg/kg), respectively, while the normoxia group was raised in a normoxic environment with daily saline injections. Right ventricular systolic pressure (RVSP) was measured using the direct pressure method. Pulmonary vascular morphology was assessed using hematoxylin and eosin staining, with metrics including the percentage of medial thickness of small pulmonary arteries relative to the external diameter (MT%) and the cross-sectional area of the media of small pulmonary arteries relative to the total cross-sectional area (MA%). Immunohistochemistry was used to detect the expression levels of hypoxia-inducible factor-1α (HIF-1α) and proliferating cell nuclear antigen (PCNA) proteins, while real-time quantitative PCR was used to to assess HIF-1α and PCNA mRNA levels. RESULTS Compared to the normoxia group, the hypoxia and hypoxia + 2ME groups showed increased RVSP and upregulated HIF-1α and PCNA protein and mRNA expression levels at 3, 7, 14, and 21 days after hypoxia (P<0.05). Furthermore, at 7, 14, and 21 days after hypoxia, the hypoxia group showed increased MT% and MA% (P<0.05). In comparison to the hypoxia group, the hypoxia + 2ME group exhibited reduced RVSP and downregulated HIF-1α and PCNA protein and mRNA expression levels, along with decreased MT% and MA% at 7, 14, and 21 days after hypoxia (P<0.05). CONCLUSIONS 2ME may protect against HPH in neonatal rats by inhibiting the expression of HIF-1α and PCNA and reducing pulmonary vascular remodeling. Citation:Chinese Journal of Contemporary Pediatrics, 2024, 26(7): 757-764.
Collapse
Affiliation(s)
- Ou Xie
- Department of Neonatology, First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, China
| | - Shan-Shan Li
- Department of Neonatology, First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, China
| | - Yang Luo
- Department of Neonatology, First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, China
| | | |
Collapse
|
4
|
Dignam JP, Sharma S, Stasinopoulos I, MacLean MR. Pulmonary arterial hypertension: Sex matters. Br J Pharmacol 2024; 181:938-966. [PMID: 37939796 DOI: 10.1111/bph.16277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 10/10/2023] [Accepted: 10/11/2023] [Indexed: 11/10/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a complex disease of multifactorial origin. While registries have demonstrated that women are more susceptible to the disease, females with PAH have superior right ventricle (RV) function and a better prognosis than their male counterparts, a phenomenon referred to as the 'estrogen paradox'. Numerous pre-clinical studies have investigated the involvement of sex hormones in PAH pathobiology, often with conflicting results. However, recent advances suggest that abnormal estrogen synthesis, metabolism and signalling underpin the sexual dimorphism of this disease. Other sex hormones, such as progesterone, testosterone and dehydroepiandrosterone may also play a role. Several non-hormonal factor including sex chromosomes and epigenetics have also been implicated. Though the underlying pathophysiological mechanisms are complex, several compounds that modulate sex hormones levels and signalling are under investigation in PAH patients. Further elucidation of the estrogen paradox will set the stage for the identification of additional therapeutic targets for this disease.
Collapse
Affiliation(s)
- Joshua P Dignam
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, Scotland, UK
| | - Smriti Sharma
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, Scotland, UK
| | - Ioannis Stasinopoulos
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, Scotland, UK
- Mass Spectrometry Core, Edinburgh Clinical Research Facility, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, Scotland, UK
| | - Margaret R MacLean
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, Scotland, UK
| |
Collapse
|
5
|
Shima N, Yamamura A, Fujiwara M, Amano T, Matsumoto K, Sekine T, Okano H, Kondo R, Suzuki Y, Yamamura H. Up-regulated expression of two-pore domain K + channels, KCNK1 and KCNK2, is involved in the proliferation and migration of pulmonary arterial smooth muscle cells in pulmonary arterial hypertension. Front Cardiovasc Med 2024; 11:1343804. [PMID: 38410243 PMCID: PMC10894933 DOI: 10.3389/fcvm.2024.1343804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 01/29/2024] [Indexed: 02/28/2024] Open
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.
Collapse
Affiliation(s)
- Natsumi Shima
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| | - Aya Yamamura
- Department of Physiology, Aichi Medical University, Nagakute, Japan
| | - Moe Fujiwara
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| | - Taiki Amano
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| | - Kazuyuki Matsumoto
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| | - Taiga Sekine
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| | - Haruka Okano
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| | - Rubii Kondo
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| | - Yoshiaki Suzuki
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| | - Hisao Yamamura
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| |
Collapse
|
6
|
郭 鑫, 李 明, 巴依尔才次克, 杨 延, 王 乐. [Effect of platelet-derived growth factor-BB on pulmonary vascular remodeling in neonatal rats with hypoxic pulmonary hypertension and its mechanism]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2023; 25:407-414. [PMID: 37073847 PMCID: PMC10120343 DOI: 10.7499/j.issn.1008-8830.2212002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 03/07/2023] [Indexed: 04/20/2023]
Abstract
OBJECTIVES To study the effect of platelet-derived growth factor-BB (PDGF-BB) on pulmonary vascular remodeling in neonatal rats with hypoxic pulmonary hypertension (HPH). METHODS A total of 128 neonatal rats were randomly divided into four groups: PDGF-BB+HPH, HPH, PDGF-BB+normal oxygen, and normal oxygen (n=32 each). The rats in the PDGF-BB+HPH and PDGF-BB+normal oxygen groups were given an injection of 13 μL 6×1010 PFU/mL adenovirus with PDGF-BB genevia the caudal vein. After 24 hours of adenovirus transfection, the rats in the HPH and PDGF-BB+HPH groups were used to establish a neonatal rat model of HPH. Right ventricular systolic pressure (RVSP) was measured on days 3, 7, 14, and 21 of hypoxia. Hematoxylin-eosin staining was used to observe pulmonary vascular morphological changes under an optical microscope, and vascular remodeling parameters (MA% and MT%) were also measured. Immunohistochemistry was used to measure the expression levels of PDGF-BB and proliferating cell nuclear antigen (PCNA) in lung tissue. RESULTS The rats in the PDGF-BB+HPH and HPH groups had a significantly higher RVSP than those of the same age in the normal oxygen group at each time point (P<0.05). The rats in the PDGF-BB+HPH group showed vascular remodeling on day 3 of hypoxia, while those in the HPH showed vascular remodeling on day 7 of hypoxia. On day 3 of hypoxia, the PDGF-BB+HPH group had significantly higher MA% and MT% than the HPH, PDGF-BB+normal oxygen, and normal oxygen groups (P<0.05). On days 7, 14, and 21 of hypoxia, the PDGF-BB+HPH and HPH groups had significantly higher MA% and MT% than the PDGF-BB+normal oxygen and normal oxygen groups (P<0.05). The PDGF-BB+HPH and HPH groups had significantly higher expression levels of PDGF-BB and PCNA than the normal oxygen group at all time points (P<0.05). On days 3, 7, and 14 of hypoxia, the PDGF-BB+HPH group had significantly higher expression levels of PDGF-BB and PCNA than the HPH group (P<0.05), while the PDGF-BB+normal oxygen group had significantly higher expression levels of PDGF-BB and PCNA than the normal oxygen group (P<0.05). CONCLUSIONS Exogenous administration of PDGF-BB in neonatal rats with HPH may upregulate the expression of PCNA, promote pulmonary vascular remodeling, and increase pulmonary artery pressure.
Collapse
Affiliation(s)
| | - 明霞 李
- 新疆医科大学第一附属医院新生儿科,新疆乌鲁木齐830054
| | - 巴依尔才次克
- 新疆医科大学第一附属医院新生儿科,新疆乌鲁木齐830054
| | | | - 乐 王
- 新疆医科大学第一附属医院新生儿科,新疆乌鲁木齐830054
| |
Collapse
|
7
|
New Drugs and Therapies in Pulmonary Arterial Hypertension. Int J Mol Sci 2023; 24:ijms24065850. [PMID: 36982922 PMCID: PMC10058689 DOI: 10.3390/ijms24065850] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 03/12/2023] [Accepted: 03/14/2023] [Indexed: 03/22/2023] Open
Abstract
Pulmonary arterial hypertension is a chronic, progressive disorder of the pulmonary vasculature with associated pulmonary and cardiac remodeling. PAH was a uniformly fatal disease until the late 1970s, but with the advent of targeted therapies, the life expectancy of patients with PAH has now considerably improved. Despite these advances, PAH inevitably remains a progressive disease with significant morbidity and mortality. Thus, there is still an unmet need for the development of new drugs and other interventional therapies for the treatment of PAH. One shortcoming of currently approved vasodilator therapies is that they do not target or reverse the underlying pathogenesis of the disease process itself. A large body of evidence has evolved in the past two decades clarifying the role of genetics, dysregulation of growth factors, inflammatory pathways, mitochondrial dysfunction, DNA damage, sex hormones, neurohormonal pathways, and iron deficiency in the pathogenesis of PAH. This review focuses on newer targets and drugs that modify these pathways as well as novel interventional therapies in PAH.
Collapse
|
8
|
Wang J, Uddin MN, Wang R, Gong YH, Wu Y. Comprehensive analysis and validation of novel immune and vascular remodeling related genes signature associated with drug interactions in pulmonary arterial hypertension. Front Genet 2022; 13:922213. [PMID: 36147486 PMCID: PMC9486302 DOI: 10.3389/fgene.2022.922213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Accepted: 08/15/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Previous studies revealed that the gene signatures are associated with the modulation and pathogenesis of pulmonary arterial hypertension (PAH). However, identifying critical transcriptional signatures in the blood of PAH patients remains lacking. Methods: The differentially expressed transcriptional signatures in the blood of PAH patients were identified by a meta-analysis from four microarray datasets. Then we investigated the enrichment of gene ontology and KEGG pathways and identified top hub genes. Besides, we investigated the correlation of crucial hub genes with immune infiltrations, hallmark gene sets, and blood vessel remodeling genes. Furthermore, we investigated the diagnostic efficacy of essential hub genes and their expression validation in an independent cohort of PAH, and we validate the expression level of hub genes in monocrotaline (MCT) induced PAH rats' model. Finally, we have identified the FDA-approved drugs that target the hub genes and their molecular docking. Results: We found 1,216 differentially expressed genes (DEGs), including 521 up-regulated and 695 down-regulated genes, in the blood of the PAH patients. The up-regulated DEGs are significantly associated with the enrichment of KEGG pathways mainly involved with immune regulation, cellular signaling, and metabolisms. We identified 13 master transcriptional regulators targeting the dysregulated genes in PAH. The STRING-based investigation identified the function of hub genes associated with multiple immune-related pathways in PAH. The expression levels of RPS27A, MAPK1, STAT1, RPS6, FBL, RPS3, RPS2, and GART are positively correlated with ssGSEA scores of various immune cells as positively correlated with the hallmark of oxidative stress. Besides, we found that these hub genes also regulate the vascular remodeling in PAH. Furthermore, the expression levels of identified hub genes showed good diagnostic efficacy in the blood of PAH, and we validated most of the hub genes are consistently dysregulated in an independent PAH cohort. Validation of hub genes expression level in the monocrotaline (MCT)-induced lung tissue of rats with PAH revealed that 5 screened hub genes (MAPK1, STAT1, TLR4, TLR2, GART) are significantly highly expressed in PAH rats, and 4 screened hub genes (RPS6, FBL, RPS3, and RPS2) are substantially lowly expressed in rats with PAH. Finally, we analyzed the interaction of hub proteins and FDA-approved drugs and revealed their molecular docking, and the results showed that MAPK1, TLR4, and GART interact with various drugs with appropriate binding affinity. Conclusion: The identified blood-derived key transcriptional signatures significantly correlate with immune infiltrations, hypoxia, glycolysis, and blood vessel remodeling genes. These findings may provide new insight into the diagnosis and treatment of PAH patients.
Collapse
Affiliation(s)
- Jie Wang
- Department of Pharmacy, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Md Nazim Uddin
- Institute of Food Science and Technology, Bangladesh Council of Scientific and Industrial Research (BCSIR), Dhaka, Bangladesh
| | - Rui Wang
- Department of General Medicine, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Yue-Hong Gong
- Department of Pharmacy, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Yun Wu
- Department of General Medicine, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| |
Collapse
|
9
|
Mickael C, Kheyfets VO, Langouët-Astrié C, Lee MH, Sanders LA, Trentin CO, Sweatt AJ, Zamanian RT, Bull TM, Stenmark K, Graham BB, Tuder RM. Peripheral Blood Inflammation Profile of Patients with Pulmonary Arterial Hypertension Using the High-Throughput Olink Proteomics Platform. Am J Respir Cell Mol Biol 2022; 66:580-581. [PMID: 35486078 PMCID: PMC9116364 DOI: 10.1165/rcmb.2021-0369le] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
| | | | | | - Michael H. Lee
- University of California, San FranciscoSan Francisco, California
| | | | | | | | | | | | | | - Brian B. Graham
- University of California, San FranciscoSan Francisco, California
| | | |
Collapse
|
10
|
Cardiovascular, Brain, and Lung Involvement in a Newborn With a Novel FLNA Mutation: A Case Report and Literature Review. Adv Neonatal Care 2022; 22:125-131. [PMID: 33852449 DOI: 10.1097/anc.0000000000000878] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Filamin A (FLNA) is an intracellular actin-binding protein, encoded by the FLNA gene, with a wide tissue expression. It is involved in several cellular functions, and extracellular matrix structuring. FLNA gene alterations lead to diseases with a wide phenotypic spectrum, such as brain periventricular nodular heterotopia (PVNH), cardiovascular abnormalities, skeletal dysplasia, and lung involvement. CLINICAL FINDINGS We present the case of a female infant who showed at birth aortic valve stenosis and PVNH, and subsequently developed interstitial lung disease with severe pulmonary hypertension. PRIMARY DIAGNOSIS The association of aortic valve dysplasia, left ventricular outflow obstruction, persistent patent ductus arteriosus, and brain heterotopic gray matter suggested a possible FLNA gene alteration. A novel heterozygous intronic variant in the FLNA gene (NM_001110556.1), c.4304-1G >A, was detected. INTERVENTIONS In consideration of valve morphology and severity of stenosis, the neonate was scheduled for a transcatheter aortic valvuloplasty. At 3 months of life, she developed hypoxemic respiratory failure with evidence of severe pulmonary hypertension. Inhaled nitric oxide (iNO) and milrinone on continuous infusion were started. Because of a partial response to iNO, an intravenous continuous infusion of sildenafil was introduced. OUTCOMES In consideration of severe clinical course and fatal outcome, the new FLNA gene mutation described in our patient seems to be associated with a loss of function of FLNA. PRACTICE RECOMMENDATIONS Lung and brain involvement, in association with left ventricular outflow obstruction and persistent patency of ductus arteriosus, should be considered highly suggestive of FLNA gene alterations, in a female newborn.
Collapse
|
11
|
Zhu Y, Sun Y, Zhang S, Li C, Zhao Y, Zhao B, Li G. Xinmai 'an extract enhances the efficacy of sildenafil in the treatment of pulmonary arterial hypertension via inhibiting MAPK signalling pathway. PHARMACEUTICAL BIOLOGY 2021; 59:594-605. [PMID: 34010580 PMCID: PMC8143608 DOI: 10.1080/13880209.2021.1917629] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 03/26/2021] [Accepted: 04/10/2021] [Indexed: 06/01/2023]
Abstract
CONTEXT Xinmai 'an tablet has been used to improve myocardial blood supply. Recently, some compounds from its formula have shown that they can treat pulmonary arterial hypertension (PAH). OBJECTIVE This study investigates the effects of Xinmai 'an extract (XMA) on PAH and further tests the co-therapeutic enhancement with sildenafil (SIL). MATERIALS AND METHODS Pulmonary artery smooth muscle cells were subjected to stimulation with SIL (12.5 μM) and XMA (250 μg/mL) for 48 h. Sprague-Dawley rats were randomly grouped into eight groups (n = 8 per group): (I) control group received saline; (II) MCT group received MCT (60 mg/kg); (III) SIL-Low group received MCT + SIL at 10 mg/kg/day; (IV) SIL-high group received MCT + SIL at 30 mg/kg/day; (V) XMA-High group received MCT + XMA at 251.6 mg/kg/day; (VI) SIL (Low)+XMA (Low) group received SIL (10 mg/kg) + XMA at 62.9 mg/kg/day; (VII) SIL (Low)+XMA (Medium) group received SIL (10 mg/kg) + XMA at 125.8 mg/kg/day; (VIII) SIL (Low)+XMA (High) group received SIL (10 mg/kg) + XMA at 251.6 mg/kg/day. Both XMA and SIL were given by gavage and were maintained daily for 2 weeks. RESULTS XMA could improve SIL's efficacy in the treatment of PAH by decreasing cell viability more effectively at non-cytotoxic concentrations (250 μg/mL) and reducing Right Ventricular Systolic Pressure (RVSP) in PAH rat. Potential mechanisms might at least in part be through activating the MAPK signalling pathway. DISCUSSION AND CONCLUSIONS The combination of XMA and SIL can improve the efficacy of pulmonary hypertension and reduce the dosage of SIL.
Collapse
Affiliation(s)
- Yaolu Zhu
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Rational Medication Evaluation and Drug Delivery Technology Lab, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yabin Sun
- Modern Chinese Medicine Institute, Hutchison Whampoa Guangzhou Baiyunshan Chinese Medicine Company Limited, Guangzhou, China
| | - Shichang Zhang
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Rational Medication Evaluation and Drug Delivery Technology Lab, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chuyuan Li
- Office of the General Manager, Hutchison Whampoa Guangzhou Baiyunshan Chinese Medicine Company Limited, Guangzhou, China
| | - Yiwei Zhao
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Rational Medication Evaluation and Drug Delivery Technology Lab, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Boxin Zhao
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Rational Medication Evaluation and Drug Delivery Technology Lab, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Guofeng Li
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Rational Medication Evaluation and Drug Delivery Technology Lab, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou, China
| |
Collapse
|
12
|
Manning EP, Ramachandra AB, Schupp JC, Cavinato C, Raredon MSB, Bärnthaler T, Cosme C, Singh I, Tellides G, Kaminski N, Humphrey JD. Mechanisms of Hypoxia-Induced Pulmonary Arterial Stiffening in Mice Revealed by a Functional Genetics Assay of Structural, Functional, and Transcriptomic Data. Front Physiol 2021; 12:726253. [PMID: 34594238 PMCID: PMC8478173 DOI: 10.3389/fphys.2021.726253] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 08/19/2021] [Indexed: 01/08/2023] Open
Abstract
Hypoxia adversely affects the pulmonary circulation of mammals, including vasoconstriction leading to elevated pulmonary arterial pressures. The clinical importance of changes in the structure and function of the large, elastic pulmonary arteries is gaining increased attention, particularly regarding impact in multiple chronic cardiopulmonary conditions. We establish a multi-disciplinary workflow to understand better transcriptional, microstructural, and functional changes of the pulmonary artery in response to sustained hypoxia and how these changes inter-relate. We exposed adult male C57BL/6J mice to normoxic or hypoxic (FiO2 10%) conditions. Excised pulmonary arteries were profiled transcriptionally using single cell RNA sequencing, imaged with multiphoton microscopy to determine microstructural features under in vivo relevant multiaxial loading, and phenotyped biomechanically to quantify associated changes in material stiffness and vasoactive capacity. Pulmonary arteries of hypoxic mice exhibited an increased material stiffness that was likely due to collagen remodeling rather than excessive deposition (fibrosis), a change in smooth muscle cell phenotype reflected by decreased contractility and altered orientation aligning these cells in the same direction as the remodeled collagen fibers, endothelial proliferation likely representing endothelial-to-mesenchymal transitioning, and a network of cell-type specific transcriptomic changes that drove these changes. These many changes resulted in a system-level increase in pulmonary arterial pulse wave velocity, which may drive a positive feedback loop exacerbating all changes. These findings demonstrate the power of a multi-scale genetic-functional assay. They also highlight the need for systems-level analyses to determine which of the many changes are clinically significant and may be potential therapeutic targets.
Collapse
Affiliation(s)
- Edward P Manning
- Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT, United States.,VA Connecticut Healthcare System, West Haven, CT, United States
| | - Abhay B Ramachandra
- Department of Biomedical Engineering, Yale University, New Haven, CT, United States
| | - Jonas C Schupp
- Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT, United States.,Respiratory Medicine, Hannover Medical School, Hannover, Germany
| | - Cristina Cavinato
- Department of Biomedical Engineering, Yale University, New Haven, CT, United States
| | - Micha Sam Brickman Raredon
- Department of Biomedical Engineering, Yale University, New Haven, CT, United States.,Vascular Biology and Therapeutics Program, Yale University, New Haven, CT, United States.,Department of Anesthesiology, Yale School of Medicine, New Haven, CT, United States
| | - Thomas Bärnthaler
- Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT, United States.,Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Carlos Cosme
- Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT, United States
| | - Inderjit Singh
- Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT, United States
| | - George Tellides
- VA Connecticut Healthcare System, West Haven, CT, United States.,Vascular Biology and Therapeutics Program, Yale University, New Haven, CT, United States.,Department of Surgery, Yale School of Medicine, New Haven, CT, United States
| | - Naftali Kaminski
- Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT, United States
| | - Jay D Humphrey
- Department of Biomedical Engineering, Yale University, New Haven, CT, United States.,Vascular Biology and Therapeutics Program, Yale University, New Haven, CT, United States
| |
Collapse
|
13
|
Rodriguez M, Chen J, Jain PP, Babicheva A, Xiong M, Li J, Lai N, Zhao T, Hernandez M, Balistrieri A, Parmisano S, Simonson T, Breen E, Valdez-Jasso D, Thistlethwaite PA, Shyy JYJ, Wang J, Garcia JGN, Makino A, Yuan JXJ. Upregulation of Calcium Homeostasis Modulators in Contractile-To-Proliferative Phenotypical Transition of Pulmonary Arterial Smooth Muscle Cells. Front Physiol 2021; 12:714785. [PMID: 34408668 PMCID: PMC8364962 DOI: 10.3389/fphys.2021.714785] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 07/13/2021] [Indexed: 12/14/2022] Open
Abstract
Excessive pulmonary artery (PA) smooth muscle cell (PASMC) proliferation and migration are implicated in the development of pathogenic pulmonary vascular remodeling characterized by concentric arterial wall thickening and arteriole muscularization in patients with pulmonary arterial hypertension (PAH). Pulmonary artery smooth muscle cell contractile-to-proliferative phenotypical transition is a process that promotes pulmonary vascular remodeling. A rise in cytosolic Ca2+ concentration [(Ca2+) cyt ] in PASMCs is a trigger for pulmonary vasoconstriction and a stimulus for pulmonary vascular remodeling. Here, we report that the calcium homeostasis modulator (CALHM), a Ca2+ (and ATP) channel that is allosterically regulated by voltage and extracellular Ca2+, is upregulated during the PASMC contractile-to-proliferative phenotypical transition. Protein expression of CALHM1/2 in primary cultured PASMCs in media containing serum and growth factors (proliferative PASMC) was significantly greater than in freshly isolated PA (contractile PASMC) from the same rat. Upregulated CALHM1/2 in proliferative PASMCs were associated with an increased ratio of pAKT/AKT and pmTOR/mTOR and an increased expression of the cell proliferation marker PCNA, whereas serum starvation and rapamycin significantly downregulated CALHM1/2. Furthermore, CALHM1/2 were upregulated in freshly isolated PA from rats with monocrotaline (MCT)-induced PH and in primary cultured PASMC from patients with PAH in comparison to normal controls. Intraperitoneal injection of CGP 37157 (0.6 mg/kg, q8H), a non-selective blocker of CALHM channels, partially reversed established experimental PH. These data suggest that CALHM upregulation is involved in PASMC contractile-to-proliferative phenotypical transition. Ca2+ influx through upregulated CALHM1/2 may play an important role in the transition of sustained vasoconstriction to excessive vascular remodeling in PAH or precapillary PH. Calcium homeostasis modulator could potentially be a target to develop novel therapies for PAH.
Collapse
Affiliation(s)
- Marisela Rodriguez
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, La Jolla, CA, United States
- Department of Pediatrics, Tucson, AZ, United States
| | - Jiyuan Chen
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, La Jolla, CA, United States
- State Key Laboratory of Respiratory Diseases, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Pritesh P. Jain
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, La Jolla, CA, United States
| | - Aleksandra Babicheva
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, La Jolla, CA, United States
| | - Mingmei Xiong
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, La Jolla, CA, United States
- State Key Laboratory of Respiratory Diseases, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jifeng Li
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, La Jolla, CA, United States
- Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Ning Lai
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, La Jolla, CA, United States
- State Key Laboratory of Respiratory Diseases, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Tengteng Zhao
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, La Jolla, CA, United States
| | - Moises Hernandez
- Division of Cardiothoracic Surgery, Department of Surgery, La Jolla, CA, United States
| | - Angela Balistrieri
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, La Jolla, CA, United States
| | - Sophia Parmisano
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, La Jolla, CA, United States
| | - Tatum Simonson
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, La Jolla, CA, United States
| | - Ellen Breen
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, La Jolla, CA, United States
| | - Daniela Valdez-Jasso
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, United States
| | | | - John Y. -J. Shyy
- Division of Cardiovascular Medicine, Department of Medicine, La Jolla, CA, United States
| | - Jian Wang
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, La Jolla, CA, United States
- State Key Laboratory of Respiratory Diseases, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Joe G. N. Garcia
- Department of Medicine, The University of Arizona, Tucson, AZ, United States
| | - Ayako Makino
- Division of Endocrinology and Metabolism, La Jolla, CA, United States
| | - Jason X. -J. Yuan
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, La Jolla, CA, United States
| |
Collapse
|
14
|
Ueland T, Abraityte A, Norum H, Varathalingam S, Gullestad L, Aukrust P, Andreassen AK. Circulating regulators of the wingless pathway in precapillary pulmonary hypertension. Respirology 2021; 26:574-581. [PMID: 33830565 DOI: 10.1111/resp.14048] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 02/18/2021] [Indexed: 12/21/2022]
Abstract
BACKGROUND AND OBJECTIVE Dysregulated Wnt signalling has been implicated in pulmonary hypertension (PH). We hypothesized that plasma levels of secreted Wnt proteins would be increased in patients with precapillary PH, correlate with indices of vascular resistance and cardiac function and give information on long-term prognosis. METHODS We measured the Wnt ligand Wnt5a and secreted Wnt antagonists Dickkopf (DKK) DKK1, DKK3, secreted frizzled-related protein 3 (sFRP3), Wnt inhibitory factor-1 (WIF1) and sclerostin (SOST) in 106 patients with precapillary PH and 40 healthy controls. A second sample was obtained after a median of 4 months (n = 52). During a median of 90 months follow-up, 67 patients died. RESULTS Our main findings were (i) Precapillary PH is characterized by enhanced systemic Wnt activity as reflected by elevated plasma levels of Wnt5a and secreted antagonists irrespective of diagnostic subgroups. (ii) WIF1 and in particular Wnt5a correlated with pulmonary vascular resistance and cardiac dysfunction. (iii) High levels of Wnt5a, sFRP3, DKK3 and WIF1 were associated with poor prognosis in age- and sex-adjusted analysis (hazard ratios per log/SD change ~1.4) and for DKK3 after further adjustment with right arterial pressure, pulmonary oxygen saturation, cardiac index, N-terminal pro B-type natriuretic peptide and peak oxygen uptake (VO2 ). Finally, an elevation of Wnt5a and DKK3 during follow-up was independently associated with poor prognosis. CONCLUSION Our data indicate that Wnt signalling pathways could be implicated in the pathogenesis of precapillary PH, and that some of the Wnt-related molecules (i.e., Wnt5a and DKK3) should be further investigated in these patients.
Collapse
Affiliation(s)
- Thor Ueland
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Faculty of Medicine, University of Oslo, Oslo, Norway.,K. G. Jebsen Thrombosis Research and Expertise Center, University of Tromsø, Tromsø, Norway
| | - Aurelija Abraityte
- Faculty of Medicine, University of Oslo, Oslo, Norway.,Center for Heart Failure Research, University of Oslo, Oslo, Norway
| | - Hilde Norum
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Department of Research and Development, Division of Emergencies and Critical Care, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Sharanga Varathalingam
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Lars Gullestad
- Faculty of Medicine, University of Oslo, Oslo, Norway.,Center for Heart Failure Research, University of Oslo, Oslo, Norway.,Department of Cardiology, Oslo University Hospital Rikshospitalet, Oslo, Norway.,K. G. Jebsen Cardiac Research Center, University of Oslo, Oslo, Norway
| | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Faculty of Medicine, University of Oslo, Oslo, Norway.,K. G. Jebsen Thrombosis Research and Expertise Center, University of Tromsø, Tromsø, Norway.,Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway.,K. G. Jebsen Inflammation Research Center, University of Oslo, Oslo, Norway
| | - Arne K Andreassen
- Department of Cardiology, Oslo University Hospital Rikshospitalet, Oslo, Norway
| |
Collapse
|
15
|
Summers ME, Richmond BW, Menon S, Sheridan RM, Kropski JA, Majka SA, Taketo MM, Bastarache JA, West JD, De Langhe S, Geraghty P, Klemm DJ, Chu HW, Friedman RS, Tao YK, Foronjy RF, Majka SM. Resident mesenchymal vascular progenitors modulate adaptive angiogenesis and pulmonary remodeling via regulation of canonical Wnt signaling. FASEB J 2020; 34:10267-10285. [PMID: 32533805 PMCID: PMC7496763 DOI: 10.1096/fj.202000629r] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 05/18/2020] [Accepted: 05/20/2020] [Indexed: 12/16/2022]
Abstract
Adaptive angiogenesis is necessary for tissue repair, however, it may also be associated with the exacerbation of injury and development of chronic disease. In these studies, we demonstrate that lung mesenchymal vascular progenitor cells (MVPC) modulate adaptive angiogenesis via lineage trace, depletion of MVPC, and modulation of β-catenin expression. Single cell sequencing confirmed MVPC as multipotential vascular progenitors, thus, genetic depletion resulted in alveolar simplification with reduced adaptive angiogenesis. Following vascular endothelial injury, Wnt activation in MVPC was sufficient to elicit an emphysema-like phenotype characterized by increased MLI, fibrosis, and MVPC driven adaptive angiogenesis. Lastly, activation of Wnt/β-catenin signaling skewed the profile of human and murine MVPC toward an adaptive phenotype. These data suggest that lung MVPC drive angiogenesis in response to injury and regulate the microvascular niche as well as subsequent distal lung tissue architecture via Wnt signaling.
Collapse
Affiliation(s)
- Megan E. Summers
- Department of MedicineDivision of Pulmonary, Critical Care & Sleep MedicineNational Jewish HealthDenverCOUSA
| | - Bradley W. Richmond
- Department of MedicineDivision of Allergy, Pulmonary and Critical Care Medicine or CardiologyVanderbilt University Medical CenterNashvilleTNUSA
| | - Swapna Menon
- Pulmonary Vascular Research Institute KochiAnalyzeDat Consulting ServicesErnakulamIndia
| | - Ryan M. Sheridan
- Department of Biochemistry and Molecular GeneticsRNA Bioscience InitiativeUniversity of Colorado School of MedicineAuroraCOUSA
| | - Jonathan A. Kropski
- Department of MedicineDivision of Allergy, Pulmonary and Critical Care Medicine or CardiologyVanderbilt University Medical CenterNashvilleTNUSA
| | - Sarah A. Majka
- Department of MedicineDivision of Pulmonary, Critical Care & Sleep MedicineNational Jewish HealthDenverCOUSA
| | - M. Mark Taketo
- Division of Experimental TherapeuticsGraduate School of MedicineKyoto UniversityKyotoJapan
| | - Julie A. Bastarache
- Department of MedicineDivision of Allergy, Pulmonary and Critical Care Medicine or CardiologyVanderbilt University Medical CenterNashvilleTNUSA
| | - James D. West
- Department of MedicineDivision of Allergy, Pulmonary and Critical Care Medicine or CardiologyVanderbilt University Medical CenterNashvilleTNUSA
| | | | - Patrick Geraghty
- Division of Pulmonary and Critical Care MedicineSUNY Downstate Medical CenterBrooklynNYUSA
| | - Dwight J. Klemm
- Department of Medicine, Pulmonary & Critical Care MedicineUniversity of ColoradoAuroraCOUSA
- Gates Center for Regenerative Medicine and Stem Cell BiologyUniversity of ColoradoAuroraCOUSA
| | - Hong Wei Chu
- Department of MedicineDivision of Pulmonary, Critical Care & Sleep MedicineNational Jewish HealthDenverCOUSA
| | | | - Yuankai K. Tao
- Pulmonary Vascular Research Institute KochiAnalyzeDat Consulting ServicesErnakulamIndia
| | - Robert F. Foronjy
- Division of Pulmonary and Critical Care MedicineSUNY Downstate Medical CenterBrooklynNYUSA
| | - Susan M. Majka
- Department of MedicineDivision of Pulmonary, Critical Care & Sleep MedicineNational Jewish HealthDenverCOUSA
- Gates Center for Regenerative Medicine and Stem Cell BiologyUniversity of ColoradoAuroraCOUSA
- Department of Biomedical ResearchNational Jewish HealthDenverCOUSA
- Biomedical EngineeringVanderbilt UniversityNashvilleTNUSA
| |
Collapse
|
16
|
Lei S, Peng F, Li ML, Duan WB, Peng CQ, Wu SJ. LncRNA-SMILR modulates RhoA/ROCK signaling by targeting miR-141 to regulate vascular remodeling in pulmonary arterial hypertension. Am J Physiol Heart Circ Physiol 2020; 319:H377-H391. [PMID: 32559140 DOI: 10.1152/ajpheart.00717.2019] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a fatal progressive disease characterized by an increased blood pressure in the pulmonary arteries. RhoA/Rho-kinase (RhoA/ROCK) signaling activation is often associated with PAH. The purpose of this study is to investigate the role and mechanisms of long noncoding RNA (lncRNA) smooth muscle-induced lncRNA (SMILR) to activate the RhoA/ROCK pathway in PAH. SMILR, microRNA-141 (miR-141), and RhoA were identified by qRT-PCR in PAH patients' serum. 3-(4,5-Dimethylthiazolyl-2)-2,5-diphenyltetrazolium bromide (MTT), wound-healing assay, cell counting kit-8 (CCK-8) assay, and flow cytometry were performed to determine cell viability, migration, proliferation, and cell cycle in human pulmonary arterial smooth muscle cells (hPASMCs) and primary PASMCs from PAH patients. We also performed bioinformatical prediction, luciferase reporter assay, and RNA-binding protein immunoprecipitation (RIP) to assess the interaction among SMILR, miR-141, and RhoA. The RhoA/ROCK pathway and proliferation-related proteins were measured by Western blotting. Finally, we introduced the small hairpin (sh)SMILR to monocrotaline-induced PAH rat model and used the hemodynamic measurement, qRT-PCR, and immunohistochemistry to examine the therapeutic effects of shSMILR. SMILR and RhoA expression were upregulated, while miR-141 expression was downregulated in PAH patients. SMILR directly interacted with miR-141 and negatively regulated its expression. Knockdown of SMILR suppressed PASMC proliferation and migration induced by hypoxia. Furthermore, overexpression of miR-141 could inhibit the RhoA/ROCK pathway by binding to RhoA, thereby repressing cell proliferation-related signals. Knockdown of SMILR significantly inhibited the Rho/ROCK activation and vascular remodeling in monocrotaline-induced rats. Knockdown of SMILR effectively elevated miR-141 expression and in turn inhibited the RhoA/ROCK pathway to regulate vascular remodeling and reduce blood pressure in PAH.NEW & NOTEWORTHY Smooth muscle enriched long noncoding RNA (SMILR), as a long noncoding RNA (lncRNA), was increased in pulmonary arterial hypertension (PAH) patients and in vitro and in vivo models. SMILR activated RhoA/ROCK signaling by targeting miR-141 to disinhibit its downstream target RhoA. SMILR knockdown or miR-141 overexpression inhibited hypoxia-induced cell proliferation and migration via repressing RhoA/ROCK signaling in pulmonary arterial smooth muscle cells (PASMCs), which was confirmed in vivo experiments that knockdown of SMILR inhibited vascular remodeling and alleviated PAH in rats. SMILR may be a promising and novel therapeutic target for the treatment and drug development of PAH.
Collapse
Affiliation(s)
- Si Lei
- Department of Pulmonary and Critical Care Medicine, the Second Xiangya Hospital, Central South University; Research Unit of Respiratory Disease, Central South University; Hunan Centre for Evidence-based Medicine, Changsha, Hunan, China
| | - Fei Peng
- Department of Pulmonary and Critical Care Medicine, the Second Xiangya Hospital, Central South University; Research Unit of Respiratory Disease, Central South University; Hunan Centre for Evidence-based Medicine, Changsha, Hunan, China
| | - Mei-Lei Li
- Department of Pulmonary and Critical Care Medicine, the Second Xiangya Hospital, Central South University; Research Unit of Respiratory Disease, Central South University; Hunan Centre for Evidence-based Medicine, Changsha, Hunan, China
| | - Wen-Bing Duan
- Department of Pulmonary and Critical Care Medicine, the Second Xiangya Hospital, Central South University; Research Unit of Respiratory Disease, Central South University; Hunan Centre for Evidence-based Medicine, Changsha, Hunan, China
| | - Cai-Qin Peng
- Department of Pulmonary and Critical Care Medicine, the Second Xiangya Hospital, Central South University; Research Unit of Respiratory Disease, Central South University; Hunan Centre for Evidence-based Medicine, Changsha, Hunan, China
| | - Shang-Jie Wu
- Department of Pulmonary and Critical Care Medicine, the Second Xiangya Hospital, Central South University; Research Unit of Respiratory Disease, Central South University; Hunan Centre for Evidence-based Medicine, Changsha, Hunan, China
| |
Collapse
|
17
|
Qin S, Predescu DN, Patel M, Drazkowski P, Ganesh B, Predescu SA. Sex differences in the proliferation of pulmonary artery endothelial cells: implications for plexiform arteriopathy. J Cell Sci 2020; 133:133/9/jcs237776. [PMID: 32409569 DOI: 10.1242/jcs.237776] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 03/13/2020] [Indexed: 12/14/2022] Open
Abstract
The sex-biased disease pulmonary arterial hypertension (PAH) is characterized by the proliferation and overgrowth of dysfunctional pulmonary artery endothelial cells (PAECs). During inflammation associated with PAH, granzyme B cleaves intersectin-1 to produce N-terminal (EHITSN) and C-terminal (SH3A-EITSN) protein fragments. In a murine model of PAH, EHITSN triggers plexiform arteriopathy via p38-ELK1-c-Fos signaling. The SH3A-EITSN fragment also influences signaling, having dominant-negative effects on ERK1 and ERK2 (also known as MAPK3 and MAPK1, respectively). Using PAECs engineered to express tagged versions of EHITSN and SH3A-EITSN, we demonstrate that the two ITSN fragments increase both p38-ELK1 activation and the ratio of p38 to ERK1 and ERK2 activity, leading to PAEC proliferation, with female cells being more responsive than male cells. Furthermore, expression of EHITSN substantially upregulates the expression and activity of the long non-coding RNA Xist in female PAECs, which in turn upregulates the X-linked gene ELK1 and represses expression of krüppel-like factor 2 (KLF2). These events are recapitulated by the PAECs of female idiopathic PAH patients, and may account for their proliferative phenotype. Thus, upregulation of Xist could be an important factor in explaining sexual dimorphism in the proliferative response of PAECs and the imbalanced sex ratio of PAH.
Collapse
Affiliation(s)
- Shanshan Qin
- Department of Internal Medicine, Pulmonary, Critical Care and Sleep Medicine, Rush University Medical Center, Chicago, IL 60612, USA
| | - Dan N Predescu
- Department of Internal Medicine, Pulmonary, Critical Care and Sleep Medicine, Rush University Medical Center, Chicago, IL 60612, USA
| | - Monal Patel
- Division of Hematology/Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Patrick Drazkowski
- Department of Internal Medicine, Pulmonary, Critical Care and Sleep Medicine, Rush University Medical Center, Chicago, IL 60612, USA
| | - Balaji Ganesh
- Division of Bioanalytics, Biophysics and Cytomics, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Sanda A Predescu
- Department of Internal Medicine, Pulmonary, Critical Care and Sleep Medicine, Rush University Medical Center, Chicago, IL 60612, USA
| |
Collapse
|
18
|
Zahid KR, Raza U, Chen J, Raj UJ, Gou D. Pathobiology of pulmonary artery hypertension: role of long non-coding RNAs. Cardiovasc Res 2020; 116:1937-1947. [PMID: 32109276 DOI: 10.1093/cvr/cvaa050] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 11/30/2019] [Accepted: 02/25/2020] [Indexed: 12/30/2022] Open
Abstract
Abstract
Pulmonary arterial hypertension (PAH) is a disease with complex pathobiology, significant morbidity and mortality, and remains without a cure. It is characterized by vascular remodelling associated with uncontrolled proliferation of pulmonary artery smooth muscle cells, endothelial cell proliferation and dysfunction, and endothelial-to-mesenchymal transition, leading to narrowing of the vascular lumen, increased vascular resistance and pulmonary arterial pressure, which inevitably results in right heart failure and death. There are multiple molecules and signalling pathways that are involved in the vascular remodelling, including non-coding RNAs, i.e. microRNAs and long non-coding RNAs (lncRNAs). It is only in recent years that the role of lncRNAs in the pathobiology of pulmonary vascular remodelling and right ventricular dysfunction is being vigorously investigated. In this review, we have summarized the current state of knowledge about the role of lncRNAs as key drivers and gatekeepers in regulating major cellular and molecular trafficking involved in the pathogenesis of PAH. In addition, we have discussed the limitations and challenges in translating lncRNA research in vivo and in therapeutic applications of lncRNAs in PAH.
Collapse
Affiliation(s)
- Kashif Rafiq Zahid
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Carson International Cancer Center, Shenzhen University, Nanhai Road, Shenzhen, Guangdong 518060, China
- Key Laboratory of Optoelectronic Devices, Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen, Guangdong 518060, China
| | - Umar Raza
- Department of Biological Sciences, National University of Medical Sciences (NUMS), Khadim Abid Majeed Road, Rawalpindi, Pakistan
| | - Jidong Chen
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Carson International Cancer Center, Shenzhen University, Nanhai Road, Shenzhen, Guangdong 518060, China
| | - Usha J Raj
- Department of Pediatrics, University of Illinois at Chicago, Chicago, IL, USA
| | - Deming Gou
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Carson International Cancer Center, Shenzhen University, Nanhai Road, Shenzhen, Guangdong 518060, China
| |
Collapse
|
19
|
Yang L, Liang H, Meng X, Shen L, Guan Z, Hei B, Yu H, Qi S, Wen X. mmu_circ_0000790 Is Involved in Pulmonary Vascular Remodeling in Mice with HPH via MicroRNA-374c-Mediated FOXC1. MOLECULAR THERAPY. NUCLEIC ACIDS 2020; 20:292-307. [PMID: 32199127 PMCID: PMC7082500 DOI: 10.1016/j.omtn.2019.12.027] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Revised: 12/23/2019] [Accepted: 12/23/2019] [Indexed: 01/05/2023]
Abstract
Recently, the identification of several circular RNAs (circRNAs) as vital regulators of microRNAs (miRNAs) underlines the increasing complexity of non-coding RNA (ncRNA)-mediated regulatory networks. This study aimed to explore the effects of mmu_circ_0000790 on the biological behaviors of pulmonary artery smooth muscle cells (PASMCs) in hypoxic pulmonary hypertension (HPH). The HPH mouse model and hypoxia-induced PASMC model were initially established, and the expression of mmu_circ_0000790 in the pulmonary vascular tissues and hypoxic PASMCs was determined using quantitative reverse transcriptase polymerase chain reaction (qRT-PCR). A series of in vitro experiments such as dual-luciferase, RNA pull-down, and RNA-binding protein immunoprecipitation (RIP) assays were conducted to evaluate the interactions among mmu_circ_0000790, microRNA-374c (miR-374c), and forkhead transcription factor 1 (FOXC1). The potential physiological functions of mmu_circ_0000790, miR-374c, and FOXC1 in hypoxic PASMCs were investigated through gain- and loss-of function approaches. Upregulated mmu_circ_0000790 was found in both the HPH-pulmonary vascular tissues and hypoxic PASMCs. Additionally, mmu_circ_0000790 could competitively bind to miR-374c and consequently upregulate the target gene of miR-374c, FOXC1. It was also observed that mmu_circ_0000790 induced proliferation and inhibited apoptosis of hypoxic PASMCs, which further promoted the pulmonary vascular remodeling in mice with HPH. Therefore, we speculate that mmu_circ_0000790 may serve as a prospective target for the treatment of patients with HPH.
Collapse
Affiliation(s)
- Lei Yang
- ICU, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar 161002, P.R. China.
| | - Huan Liang
- ICU, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar 161002, P.R. China
| | - Xianguo Meng
- ICU, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar 161002, P.R. China
| | - Li Shen
- Glorious Community, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar 161002, P.R. China
| | - Zhanjiang Guan
- ICU, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar 161002, P.R. China
| | - Bingchang Hei
- ICU, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar 161002, P.R. China
| | - Haitao Yu
- ICU, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar 161002, P.R. China
| | - Shanshan Qi
- ICU, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar 161002, P.R. China
| | - Xianchun Wen
- Institute of Medical Science, Qiqihar Medical College, Qiqihar 161002, P.R. China.
| |
Collapse
|
20
|
Yan L, Cogan JD, Hedges LK, Nunley B, Hamid R, Austin ED. The Y Chromosome Regulates BMPR2 Expression via SRY: A Possible Reason "Why" Fewer Males Develop Pulmonary Arterial Hypertension. Am J Respir Crit Care Med 2019; 198:1581-1583. [PMID: 30252494 DOI: 10.1164/rccm.201802-0308le] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Affiliation(s)
- Ling Yan
- 1 Vanderbilt University Medical Center Nashville, Tennessee
| | - Joy D Cogan
- 1 Vanderbilt University Medical Center Nashville, Tennessee
| | - Lora K Hedges
- 1 Vanderbilt University Medical Center Nashville, Tennessee
| | - Bethany Nunley
- 1 Vanderbilt University Medical Center Nashville, Tennessee
| | - Rizwan Hamid
- 1 Vanderbilt University Medical Center Nashville, Tennessee
| | - Eric D Austin
- 1 Vanderbilt University Medical Center Nashville, Tennessee
| |
Collapse
|
21
|
Wang P, Zhang C, Li J, Luo L, Zhang S, Dong F, Tang Z, Ni S. Adipose-derived mesenchymal stromal cells improve hemodynamic function in pulmonary arterial hypertension: identification of microRNAs implicated in modulating endothelial function. Cytotherapy 2019; 21:416-427. [PMID: 30904330 DOI: 10.1016/j.jcyt.2019.02.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 02/23/2019] [Accepted: 02/25/2019] [Indexed: 11/18/2022]
Abstract
Pulmonary arterial hypertension (PAH) is characterized by pulmonary arterial endothelial hyperproliferation and dysfunction. Restoration of endothelial function is a common goal of available treatments. In the present study, human adipose-derived mesenchymal stromal cells (ASCs) were co-cultured with monocrotaline pyrrole-treated human pulmonary arterial endothelial cells (HPAECs); increased proliferation of HPAECs and expression of vascular endothelial growth factor (VEGF) were observed. High throughput sequencing results showed that six microRNAs (miMNAs) of ASCs were significantly dysregulated. In monocrotaline-induced PAH rat models, ASC transplantation improved the right ventricle systolic pressure, right ventricle hypertrophy and pulmonary endothelium hyperproliferation, and four of the six miRNAs were validated in the lung tissue samples. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis showed that these dysregulated miRNAs were involved in the regulation of transcription, signal transduction, negative regulation of cell proliferation through mitogen-activated protein kinase (MAPK) signaling pathway, Wnt signaling pathway, VEGF signaling pathway, cytokine-cytokine receptor interaction, regulation of actin cytoskeleton, transforming growth factor (TGF)-beta signaling pathway and P53 signaling pathway. Our data indicates that the unique six miRNA expression signature could be involved in the PAH endothelial repair by ASCs.
Collapse
Affiliation(s)
- Pengbo Wang
- Department of Respiratory and Critical Care Medicine, Affiliated hospital of Nantong University, Nantong, Jiangsu, China; Medical School of Nantong University, Nantong, Jiangsu, China
| | - Caixin Zhang
- Department of Respiratory and Critical Care Medicine, Affiliated hospital of Nantong University, Nantong, Jiangsu, China; Medical School of Nantong University, Nantong, Jiangsu, China
| | - Jun Li
- Department of Respiratory and Critical Care Medicine, Affiliated hospital of Nantong University, Nantong, Jiangsu, China; Medical School of Nantong University, Nantong, Jiangsu, China
| | - Lin Luo
- Medical School of Nantong University, Nantong, Jiangsu, China
| | - Shuwen Zhang
- Department of Respiratory and Critical Care Medicine, Affiliated hospital of Nantong University, Nantong, Jiangsu, China; Medical School of Nantong University, Nantong, Jiangsu, China
| | - Fulu Dong
- Medical School of Nantong University, Nantong, Jiangsu, China
| | - Zhiyuan Tang
- Department of Respiratory and Critical Care Medicine, Affiliated hospital of Nantong University, Nantong, Jiangsu, China; Medical School of Nantong University, Nantong, Jiangsu, China.
| | - Songshi Ni
- Department of Respiratory and Critical Care Medicine, Affiliated hospital of Nantong University, Nantong, Jiangsu, China; Medical School of Nantong University, Nantong, Jiangsu, China.
| |
Collapse
|
22
|
Docherty CK, Harvey KY, Mair KM, Griffin S, Denver N, MacLean MR. The Role of Sex in the Pathophysiology of Pulmonary Hypertension. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1065:511-528. [PMID: 30051404 DOI: 10.1007/978-3-319-77932-4_31] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a progressive disease characterised by increased pulmonary vascular resistance and pulmonary artery remodelling as result of increased vascular tone and vascular cell proliferation, respectively. Eventually, this leads to right heart failure. Heritable PAH is caused by a mutation in the bone morphogenetic protein receptor-II (BMPR-II). Female susceptibility to PAH has been known for some time, and most recent figures show a female-to-male ratio of 4:1. Variations in the female sex hormone estrogen and estrogen metabolism modify FPAH risk, and penetrance of the disease in BMPR-II mutation carriers is increased in females. Several lines of evidence point towards estrogen being pathogenic in the pulmonary circulation, and thus increasing the risk of females developing PAH. Recent studies have also suggested that estrogen metabolism may be crucial in the development and progression of PAH with studies indicating that downstream metabolites such as 16α-hydroxyestrone are upregulated in several forms of experimental pulmonary hypertension (PH) and can cause pulmonary artery smooth muscle cell proliferation and subsequent vascular remodelling. Conversely, other estrogen metabolites such as 2-methoxyestradiol have been shown to be protective in the context of PAH. Estrogen may also upregulate the signalling pathways of other key mediators of PAH such as serotonin.
Collapse
Affiliation(s)
- Craig K Docherty
- Research Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Katie Yates Harvey
- Research Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Kirsty M Mair
- Research Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Sinead Griffin
- Research Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Nina Denver
- Research Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Margaret R MacLean
- Research Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK.
| |
Collapse
|
23
|
Jiang L, Zhou X, Yang H, Guan R, Xin Y, Wang J, Shen L, Zhu D, Ma S, Wang J. Upregulation of AT 1 Receptor Mediates a Pressor Effect Through ROS-SAPK/JNK Signaling in Glutamatergic Neurons of Rostral Ventrolateral Medulla in Rats With Stress-Induced Hypertension. Front Physiol 2019; 9:1860. [PMID: 30670978 PMCID: PMC6331519 DOI: 10.3389/fphys.2018.01860] [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: 07/25/2018] [Accepted: 12/11/2018] [Indexed: 11/29/2022] Open
Abstract
The present study examined whether angiotensin II (Ang II) mediates the pressor effect through nicotinamide adenine dinucleotide phosphate (NADPH) oxidase-derived reactive oxygen species (ROS)-mitogen-activated protein kinase (MAPK) signaling in the glutamatergic neurons of the rostral ventrolateral medulla (RVLM) in stress-induced hypertensive rats (SIHR). The SIHR model was established using electric foot-shocks combined with noises for 15 days. We observed that Ang II type 1 receptor (AT1R) and the glutamatergic neurons co-localized in the RVLM of SIHR. Furthermore, glutamate levels in the intermediolateral column of the spinal cord were higher in SIHR than in controls. Microinjection of Ang II into the RVLM of SIHR activated stress-activated protein kinase/Jun N-terminal kinase (SAPK/JNK), extracellular signal-regulated protein kinase (ERK) 1/2, and p38MAPK. Compared with controls, the activation of SAPK/JNK, ERK1/2, p38MAPK, and ROS in the RVLM were higher in SIHR, an effect that was blocked by an NADPH oxidase inhibitor (apocynin) and an AT1R antagonist (candesartan). RVLM microinjection of apocynin or a SAPK/JNK inhibitor (SP600125), but not an ERK1/2 inhibitor (U0126) or a p38MAPK inhibitor (SB203580), decreased AT1R mRNA and mean arterial blood pressure (MABP) in SIHR. The increase of AT1R protein expression and MABP was inhibited by intracerebroventricular infusion (ICV), for 14 days, of SP600125, but not U0126 or SB203580 in SIHR. We conclude that Ang II modulates the pressor effect through AT1R-dependent ROS-SAPK/JNK signaling in glutamatergic neurons in the RVLM of SIHR.
Collapse
Affiliation(s)
- Liping Jiang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Xuan Zhou
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Hongyu Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Ruijuan Guan
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yanlei Xin
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Jijiang Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Linlin Shen
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Danian Zhu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Shulan Ma
- Training Center of Medical Experiments, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Jin Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
24
|
Tang H, Wu K, Wang J, Vinjamuri S, Gu Y, Song S, Wang Z, Zhang Q, Balistrieri A, Ayon RJ, Rischard F, Vanderpool R, Chen J, Zhou G, Desai AA, Black SM, Garcia JGN, Yuan JXJ, Makino A. Pathogenic Role of mTORC1 and mTORC2 in Pulmonary Hypertension. JACC Basic Transl Sci 2018; 3:744-762. [PMID: 30623134 PMCID: PMC6314964 DOI: 10.1016/j.jacbts.2018.08.009] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 06/23/2018] [Accepted: 08/16/2018] [Indexed: 01/07/2023]
Abstract
G protein-coupled receptors and tyrosine kinase receptors signal through the phosphoinositide 3-kinase/Akt/mTOR pathway to induce cell proliferation, survival, and growth. mTOR is a kinase present in 2 functionally distinct complexes, mTORC1 and mTORC2. Functional disruption of mTORC1 by knockout of Raptor (regulatory associated protein of mammalian target of rapamycin) in smooth muscle cells ameliorated the development of experimental PH. Functional disruption of mTORC2 by knockout of Rictor (rapamycin insensitive companion of mammalian target of rapamycin) caused spontaneous PH by up-regulating platelet-derived growth factor receptors. Use of mTOR inhibitors (e.g., rapamycin) to treat PH should be accompanied by inhibitors of platelet-derived growth factor receptors (e.g., imatinib).
Concentric lung vascular wall thickening due to enhanced proliferation of pulmonary arterial smooth muscle cells is an important pathological cause for the elevated pulmonary vascular resistance reported in patients with pulmonary arterial hypertension. We identified a differential role of mammalian target of rapamycin (mTOR) complex 1 and complex 2, two functionally distinct mTOR complexes, in the development of pulmonary hypertension (PH). Inhibition of mTOR complex 1 attenuated the development of PH; however, inhibition of mTOR complex 2 caused spontaneous PH, potentially due to up-regulation of platelet-derived growth factor receptors in pulmonary arterial smooth muscle cells, and compromised the therapeutic effect of the mTOR inhibitors on PH. In addition, we describe a promising therapeutic strategy using combination treatment with the mTOR inhibitors and the platelet-derived growth factor receptor inhibitors on PH and right ventricular hypertrophy. The data from this study provide an important mechanism-based perspective for developing novel therapies for patients with pulmonary arterial hypertension and right heart failure.
Collapse
Key Words
- EC, endothelial cell
- FOXO3a, Forkhead box O3a
- GPCR, G protein-coupled receptor
- HPH, hypoxia-induced pulmonary hypertension
- PA, pulmonary artery
- PAEC, pulmonary arterial endothelial cell
- PAH, pulmonary arterial hypertension
- PASMC, pulmonary arterial smooth muscle cell
- PDGF, platelet-derived growth factor
- PDGFR, platelet-derived growth factor receptor
- PH, pulmonary hypertension
- PI3K, phosphoinositide 3-kinase
- PTEN, phosphatase and tensin homolog
- PVR, pulmonary vascular resistance
- RVH, right ventricular hypertrophy
- RVSP, right ventricular systolic pressure
- Raptor
- Raptor, regulatory associated protein of mammalian target of rapamycin
- Rictor
- Rictor, rapamycin insensitive companion of mammalian target of rapamycin
- SM, smooth muscle
- TKR, tyrosine kinase receptor
- WT, wild-type
- mTOR
- mTORC1, mammalian target of rapamycin complex 1
- mTORC2, mammalian target of rapamycin complex 2
- pAKT, phosphorylated AKT
- pulmonary hypertension
- right ventricle
Collapse
Affiliation(s)
- Haiyang Tang
- Division of Translational and Regenerative Medicine, The University of Arizona College of Medicine, Tucson, Arizona.,State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Kang Wu
- Division of Translational and Regenerative Medicine, The University of Arizona College of Medicine, Tucson, Arizona.,State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jian Wang
- Division of Translational and Regenerative Medicine, The University of Arizona College of Medicine, Tucson, Arizona.,State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Sujana Vinjamuri
- Division of Translational and Regenerative Medicine, The University of Arizona College of Medicine, Tucson, Arizona
| | - Yali Gu
- Division of Translational and Regenerative Medicine, The University of Arizona College of Medicine, Tucson, Arizona
| | - Shanshan Song
- Division of Translational and Regenerative Medicine, The University of Arizona College of Medicine, Tucson, Arizona
| | - Ziyi Wang
- Division of Translational and Regenerative Medicine, The University of Arizona College of Medicine, Tucson, Arizona.,State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qian Zhang
- Division of Translational and Regenerative Medicine, The University of Arizona College of Medicine, Tucson, Arizona.,State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Department of Physiology, The University of Arizona College of Medicine, Tucson, Arizona
| | - Angela Balistrieri
- Division of Translational and Regenerative Medicine, The University of Arizona College of Medicine, Tucson, Arizona
| | - Ramon J Ayon
- Division of Translational and Regenerative Medicine, The University of Arizona College of Medicine, Tucson, Arizona
| | - Franz Rischard
- Division of Translational and Regenerative Medicine, The University of Arizona College of Medicine, Tucson, Arizona.,Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, The University of Arizona College of Medicine, Tucson, Arizona
| | - Rebecca Vanderpool
- Division of Translational and Regenerative Medicine, The University of Arizona College of Medicine, Tucson, Arizona
| | - Jiwang Chen
- Department of Pediatrics, University of Illinois College of Medicine, Chicago, Illinois
| | - Guofei Zhou
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Department of Pediatrics, University of Illinois College of Medicine, Chicago, Illinois
| | - Ankit A Desai
- Division of Translational and Regenerative Medicine, The University of Arizona College of Medicine, Tucson, Arizona.,Division of Cardiology, Department of Medicine, The University of Arizona College of Medicine, Tucson, Arizona
| | - Stephen M Black
- Division of Translational and Regenerative Medicine, The University of Arizona College of Medicine, Tucson, Arizona.,Department of Physiology, The University of Arizona College of Medicine, Tucson, Arizona
| | - Joe G N Garcia
- Division of Translational and Regenerative Medicine, The University of Arizona College of Medicine, Tucson, Arizona.,Department of Physiology, The University of Arizona College of Medicine, Tucson, Arizona.,Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, The University of Arizona College of Medicine, Tucson, Arizona
| | - Jason X-J Yuan
- Division of Translational and Regenerative Medicine, The University of Arizona College of Medicine, Tucson, Arizona.,State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Department of Physiology, The University of Arizona College of Medicine, Tucson, Arizona
| | - Ayako Makino
- Division of Translational and Regenerative Medicine, The University of Arizona College of Medicine, Tucson, Arizona.,Department of Physiology, The University of Arizona College of Medicine, Tucson, Arizona
| |
Collapse
|
25
|
Wei H, Zhang D, Liu L, Xia W, Li F. Rho signaling pathway enhances proliferation of PASMCs by suppressing nuclear translocation of Smad1 in PAH. Exp Ther Med 2018; 17:71-78. [PMID: 30603049 PMCID: PMC6307528 DOI: 10.3892/etm.2018.6942] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Accepted: 07/26/2018] [Indexed: 12/23/2022] Open
Abstract
Bone morphogenetic protein (BMP) and Rho kinase signaling pathways exert counter regulatory effects on pulmonary artery smooth muscle cell (PASMC) proliferation in pulmonary artery hypertension (PAH). To elucidate the mechanism of this interaction, the present study tested whether Rho kinase activated by platelet derived growth factor-BB (PDGF-BB) enhances PASMC proliferation by suppressing the nuclear translocation of Smad1 induced by BMP-2. BMP-2 was used to activate the Smad1 signaling pathway and PDGF-BB was used to activate the Rho kinase signaling pathway when cells were pretreated with or without Rho-associated protein kinase (ROCK) inhibitor Y-27632 or dual specificity mitogen-activated protein kinase kinase (MEK) 1 and 2 inhibitor U0126. Western blotting was used to determine the expression of the components of the Rho signaling pathway, and the expression of various variants of phosphorylated mothers against decapentaplegic homolog (p-Smad)1 in the cytoplasm and nucleus. Immunofluorescent staining was used to observe subcellular distribution of p-Smad1. A cell counting kit was used to analyze cell proliferation. Active RhoA/Rho kinase signaling and decreased nuclear translocation of Smad1 were found in primary cultured PASMCs from the rat model of PAH compared with the control PASMCs. Treatment with BMP-2 significantly increased nuclear accumulation of Smad1 and inhibited the proliferation of PASMCs. However, pretreatment with PDGF-BB significantly decreased the nuclear accumulation of Smad1 induced by BMP-2 and enhanced the proliferation of PASMCs. Furthermore, pretreatment with Y-27632 or U0126 was found to restore the nuclear translocation of Smad1 suppressed by PDGF-BB and decrease the proliferation of PASMCs. In conclusion, the present study suggested that Rho kinase activated by PDGF-BB suppressed BMP-2-induced nuclear translocation of Smad1 via the MEK/mitogen-activated protein kinase and enhanced BMP-2-inhibited proliferation of PASMCs.
Collapse
Affiliation(s)
- Hongwei Wei
- Department of Pediatrics, The Third Hospital of Jinan, Jinan, Shandong 250132, P.R. China
| | - Dongqing Zhang
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Lili Liu
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Wei Xia
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Fuhai Li
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| |
Collapse
|
26
|
Wang J, Zhu MC, Kalionis B, Wu JZ, Wang LL, Ge HY, Chen CC, Tang XD, Song YL, He H, Xia SJ. Characteristics of circular RNA expression in lung tissues from mice with hypoxia‑induced pulmonary hypertension. Int J Mol Med 2018; 42:1353-1366. [PMID: 29956720 PMCID: PMC6089758 DOI: 10.3892/ijmm.2018.3740] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Accepted: 06/20/2018] [Indexed: 02/07/2023] Open
Abstract
Pulmonary hypertension (PH) is a life-threatening lung disease, characterized by an increase in pulmonary arterial pressure caused by vasoconstriction and vascular remodeling. The pathogenesis of PH is not fully understood, and there is a lack of potential biomarkers for the diagnosis and treatment of patients with PH. Non-coding RNAs with a characteristic covalently closed loop structure, termed circular RNAs (circRNAs), are present in a number of pulmonary diseases. To the best of our knowledge, the present study is the first to use microarray analysis to determine the expression profile of circRNAs in lung tissues from mice with hypoxia-induced PH. In total, 23 significantly upregulated and 41 significantly down-regulated circRNAs were identified. Of these, 12 differentially expressed circRNAs were selected for further validation using reverse transcription-quantitative polymerase chain reaction. Putative microRNAs (miRNAs) that bind to the dysregulated circRNAs were predicted. Subsequently, bioinformatics tools were used to construct circRNA-miRNA-mRNA networks for the two most promising circRNAs, namely mmu_circRNA_004592 and mmu_circRNA_018351. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses of target genes of the dysregulated circRNAs revealed that these dysregulated circRNAs may serve an important role in the pathogenesis of hypoxia-induced PH. Therefore, these dysregulated circRNAs are candidate diagnostic biomarkers and potential therapeutic targets for PH.
Collapse
Affiliation(s)
- Jian Wang
- Department of Geriatrics, Shanghai Institute of Geriatrics, Huadong Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Meng-Chan Zhu
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai 200030, P.R. China
| | - Bill Kalionis
- Pregnancy Research Centre, Department of Maternal‑Fetal Medicine, Royal Women's Hospital, Parkville, Victoria 3052, Australia
| | - Jun-Zhen Wu
- Department of Geriatrics, Shanghai Institute of Geriatrics, Huadong Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Lin-Lin Wang
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai 200030, P.R. China
| | - Hai-Yan Ge
- Department of Pulmonary Medicine, Huadong Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Cui-Cui Chen
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai 200030, P.R. China
| | - Xiao-Dan Tang
- Department of Pulmonary Medicine, Huadong Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Yuan-Lin Song
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai 200030, P.R. China
| | - Hong He
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China
| | - Shi-Jin Xia
- Department of Geriatrics, Shanghai Institute of Geriatrics, Huadong Hospital, Fudan University, Shanghai 200040, P.R. China
| |
Collapse
|
27
|
Bessa-Gonçalves M, Bragança B, Martins-Dias E, Correia-de-Sá P, Fontes-Sousa AP. Is the adenosine A 2B 'biased' receptor a valuable target for the treatment of pulmonary arterial hypertension? Drug Discov Today 2018; 23:1285-1292. [PMID: 29747005 DOI: 10.1016/j.drudis.2018.05.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 03/25/2018] [Accepted: 05/02/2018] [Indexed: 12/12/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a maladaptive disorder characterized by increased pulmonary vascular resistance leading to right ventricular failure and death. Adenosine released by injured tissues, such as the lung and heart, influences tissue remodeling through the activation of adenosine receptors. Evidence regarding activation of the low-affinity A2BAR by adenosine points towards pivotal roles of this receptor in processes associated with both acute and chronic lung diseases. Conflicting results exist concerning the beneficial or detrimental roles of the A2B 'biased' receptor in right ventricular failure secondary to PAH. In this review, we discuss the pros and cons of manipulating A2BARs as a putative therapeutic target in PAH.
Collapse
Affiliation(s)
- Mafalda Bessa-Gonçalves
- Laboratório de Farmacologia e Neurobiologia, Centro de Investigação Farmacológica e Inovação Medicamentosa (MedInUP), Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto (ICBAS-UP), Porto, Portugal
| | - Bruno Bragança
- Laboratório de Farmacologia e Neurobiologia, Centro de Investigação Farmacológica e Inovação Medicamentosa (MedInUP), Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto (ICBAS-UP), Porto, Portugal
| | - Eduardo Martins-Dias
- Laboratório de Farmacologia e Neurobiologia, Centro de Investigação Farmacológica e Inovação Medicamentosa (MedInUP), Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto (ICBAS-UP), Porto, Portugal
| | - Paulo Correia-de-Sá
- Laboratório de Farmacologia e Neurobiologia, Centro de Investigação Farmacológica e Inovação Medicamentosa (MedInUP), Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto (ICBAS-UP), Porto, Portugal
| | - Ana Patrícia Fontes-Sousa
- Laboratório de Farmacologia e Neurobiologia, Centro de Investigação Farmacológica e Inovação Medicamentosa (MedInUP), Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto (ICBAS-UP), Porto, Portugal.
| |
Collapse
|
28
|
Batton KA, Austin CO, Bruno KA, Burger CD, Shapiro BP, Fairweather D. Sex differences in pulmonary arterial hypertension: role of infection and autoimmunity in the pathogenesis of disease. Biol Sex Differ 2018; 9:15. [PMID: 29669571 PMCID: PMC5907450 DOI: 10.1186/s13293-018-0176-8] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 04/09/2018] [Indexed: 01/14/2023] Open
Abstract
Registry data worldwide indicate an overall female predominance for pulmonary arterial hypertension (PAH) of 2–4 over men. Genetic predisposition accounts for only 1–5% of PAH cases, while autoimmune diseases and infections are closely linked to PAH. Idiopathic PAH may include patients with undiagnosed autoimmune diseases based on the relatively high presence of autoantibodies in this group. The two largest PAH registries to date report a sex ratio for autoimmune connective tissue disease-associated PAH of 9:1 female to male, highlighting the need for future studies to analyze subgroup data according to sex. Autoimmune diseases that have been associated with PAH include female-dominant systemic sclerosis, systemic lupus erythematosus, rheumatoid arthritis, Sjögren’s syndrome, and thyroiditis as well as male-dominant autoimmune diseases like myocarditis which has been linked to HIV-associated PAH. The sex-specific association of PAH to certain infections and autoimmune diseases suggests that sex hormones and inflammation may play an important role in driving the pathogenesis of disease. However, there is a paucity of data on sex differences in inflammation in PAH, and more research is needed to better understand the pathogenesis underlying PAH in men and women. This review uses data on sex differences in PAH and PAH-associated autoimmune diseases from registries to provide insight into the pathogenesis of disease.
Collapse
Affiliation(s)
- Kyle A Batton
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, USA
| | | | - Katelyn A Bruno
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, USA
| | - Charles D Burger
- Department of Pulmonary and Critical Care Medicine, Mayo Clinic, Jacksonville, FL, USA
| | - Brian P Shapiro
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, USA
| | - DeLisa Fairweather
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, USA.
| |
Collapse
|
29
|
Fu C, Li N, Yuan Y, Wang R, Chen J, Yang J, Guo Z, Wang S, Zhang Y, Liu Y, Dong J. Chronic intermittent hypobaric hypoxia provides vascular protection in the aorta of the 2-kidney, 1-clip rat model of hypertension. Can J Physiol Pharmacol 2018; 96:807-814. [PMID: 29400080 DOI: 10.1139/cjpp-2017-0356] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Many studies have demonstrated that chronic intermittent hypobaric hypoxia (CIHH) can reduce blood pressure in spontaneously hypertensive rats and renovascular hypertensive (RVH) rats in which endothelial dysfunction is determined as a critical factor. However, whether CIHH can regulate vasodilation of the aorta in RVH rats remains unknown. The purpose of this study was to investigate the effect of CIHH on impaired relaxation of the aorta in the 2-kidney, 1-clip (2K1C) RVH rat model. The results showed CIHH improved the impaired endothelium-dependent relaxation in the 2K1C rat aorta. The endothelial dysfunction was prevented by the p38 antagonist SB203580, but not by the ERK1/2 antagonist PD98059 or JNK antagonist SP600125. Furthermore, the expression of p-eNOS, HIF-1α, and HIF-2α increased while that of p-p38 and BMP-4 decreased in CIHH-treated aortas from 2K1C rats. Finally, the p-eNOS expression was upregulated and the p-p38 expression was downregulated by pre-incubation of SB203580 or the BMP-4 antagonist Noggin with the aorta. CIHH ameliorated the impairment of endothelium-dependent relaxation through upregulating the expression of p-eNOS, which may be mediated by the inhibition of BMP-4/p-p38 MAPK, and upregulating the expression of HIFs in the 2K1C rat aorta.
Collapse
Affiliation(s)
- Congrui Fu
- a Department of Physiology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Na Li
- b Department of Physiology, Medical College, Hebei University, Baoding, Hebei, China
| | - Yujia Yuan
- a Department of Physiology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Ri Wang
- a Department of Physiology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Jinting Chen
- a Department of Physiology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Jing Yang
- a Department of Physiology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Zan Guo
- a Department of Physiology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Sheng Wang
- a Department of Physiology, Hebei Medical University, Shijiazhuang, Hebei, China.,c Hebei Collaborative Innovation Center for Cardio-Cerebrovascular Disease, Shijiazhuang, Hebei, China
| | - Yi Zhang
- a Department of Physiology, Hebei Medical University, Shijiazhuang, Hebei, China.,c Hebei Collaborative Innovation Center for Cardio-Cerebrovascular Disease, Shijiazhuang, Hebei, China
| | - Yixian Liu
- a Department of Physiology, Hebei Medical University, Shijiazhuang, Hebei, China.,c Hebei Collaborative Innovation Center for Cardio-Cerebrovascular Disease, Shijiazhuang, Hebei, China
| | - Jinghui Dong
- a Department of Physiology, Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
30
|
Shi R, Wei Z, Zhu D, Fu N, Wang C, Yin S, Liang Y, Xing J, Wang X, Wang Y. Baicalein attenuates monocrotaline-induced pulmonary arterial hypertension by inhibiting vascular remodeling in rats. Pulm Pharmacol Ther 2017; 48:124-135. [PMID: 29133079 DOI: 10.1016/j.pupt.2017.11.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 11/01/2017] [Accepted: 11/09/2017] [Indexed: 12/21/2022]
Abstract
BACKGROUND Pulmonary arterial hypertension (PAH) is a devastating cardiopulmonary disorder characterized by elevated pulmonary arterial pressure (PAP) and right ventricular hypertrophy (RVH) driven by progressive vascular remodeling. Reversing adverse vascular remodeling is an important concept in the treatment of PAH. Endothelial injury, inflammation, and oxidative stress are three main contributors to pulmonary vascular remodeling. Baicalein is a natural flavonoid that has been shown to possess anti-proliferative, anti-inflammatory, anti-oxidative, and cardioprotective properties. We hypothesized that baicalein may prevent the progression of PAH and preserve the right heart function by inhibiting pulmonary arterial remodeling. METHODS Male Sprague-Dawley rats were distributed randomly into 4 groups: control, monocrotaline (MCT)-exposed, and MCT-exposed plus baicalein treated rats (50 and 100 mg/kg/day for 2 weeks). Hemodynamic changes, RVH, and lung morphological features were examined on day 28. Apoptosis was determined by TUNEL staining, and the mRNA levels of tumor necrosis factor alpha (TNF-α), interleukin-1β (IL-1β), and IL-6 were detected by qRT-PCR. The changes in oxidative indicators, including malondialdehyde (MDA), superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) were measured using corresponding commercial kits. The levels of Bax, Bcl-2, and cleaved caspase-3, and the activation of mitogen-activated protein kinase (MAPK) and NF-κB were assessed by western blotting. RESULTS MCT induced an increase in hemodynamic parameters and RVH, which were attenuated by baicalein treatment. Baicalein also blocked MCT-induced pulmonary arterial remodeling. The levels of apoptotic (Bax/Bcl-2 ratio and cleaved caspase-3) and inflammatory (IL-6, TNF-α, and IL-1β) biomarkers in lung tissue were lower in baicalein-treated groups. Baicalein also decreased MDA level, and increased SOD and GSH-Px activity in rat pulmonary tissue. Furthermore, baicalein inhibited MCT-induced activation of the MAPK and NF-κB pathways. CONCLUSION Baicalein ameliorates MCT-induced PAH by inhibiting pulmonary arterial remodeling at least partially via the MAPK and NF-κB pathways in rats.
Collapse
Affiliation(s)
- Ruizan Shi
- Department of Pharmacology, Shanxi Medical University, Taiyuan, 030001, China.
| | - Zehui Wei
- Department of Pharmacology, Shanxi Medical University, Taiyuan, 030001, China
| | - Diying Zhu
- Department of Pharmacology, Shanxi Medical University, Taiyuan, 030001, China
| | - Naijie Fu
- Department of Pharmacology, Shanxi Medical University, Taiyuan, 030001, China
| | - Chang Wang
- Department of Pharmacology, Shanxi Medical University, Taiyuan, 030001, China
| | - Sha Yin
- Department of Pharmacology, Shanxi Medical University, Taiyuan, 030001, China
| | - Yueqin Liang
- Medical Functional Experimental Center, Shanxi Medical University, Taiyuan, 030001, China
| | - Jianfeng Xing
- Medical Functional Experimental Center, Shanxi Medical University, Taiyuan, 030001, China
| | - Xuening Wang
- Department of Cardiovascular Surgery, Shanxi Academy of Medical Sciences, Shanxi Dayi Hospital, Taiyuan, 030032, China
| | - Yan Wang
- Department of Pharmacology, Shanxi Medical University, Taiyuan, 030001, China
| |
Collapse
|
31
|
Fülöp GÁ, Yabluchanskiy A. Cyp2c44-mediated decrease of 15-HETE exacerbates pulmonary hypertension. Am J Physiol Heart Circ Physiol 2017. [PMID: 28626080 DOI: 10.1152/ajpheart.00320.2017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Gábor Á Fülöp
- Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma; and.,Translational Geroscience Laboratory, Donald W. Reynolds Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Andriy Yabluchanskiy
- Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma; and .,Translational Geroscience Laboratory, Donald W. Reynolds Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| |
Collapse
|
32
|
Hashimoto R, Joshi SR, Jiang H, Capdevila JH, McMurtry IF, Laniado Schwartzman M, Gupte SA. Cyp2c44 gene disruption is associated with increased hematopoietic stem cells: implication in chronic hypoxia-induced pulmonary hypertension. Am J Physiol Heart Circ Physiol 2017; 313:H293-H303. [PMID: 28550179 DOI: 10.1152/ajpheart.00785.2016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 05/05/2017] [Accepted: 05/20/2017] [Indexed: 01/02/2023]
Abstract
We have recently demonstrated that disruption of the murine cytochrome P-450 2c44 gene (Cyp2c44) exacerbates chronic hypoxia-induced pulmonary artery remodeling and hypertension in mice. Subsequently, we serendipitously found that Cyp2c44 gene disruption also increases hematopoietic stem cell (HSC) numbers in bone marrow and blood. Therefore, the objective of the present study was to investigate whether CYP2C44-derived eicosanoids regulate HSC proliferation/cell growth and whether increased HSCs contribute to chronic hypoxia-induced remodeling of pulmonary arteries in Cyp2c44 knockout mice. Our findings demonstrated that lack of CYP2C44 epoxygenase, which catalyzed the oxidation of arachidonic acid to epoxyeicosatrienoic (EETs) and hydroxyeicosatetraenoic (HETE) acids, increases the numbers of 1) HSCs (CD34+, CD117+, and CD133+), 2) proangiogenic (CD34+CD133+ and CD34+CD117+CD133+) cells, and 3) immunogenic/inflammatory (CD34+CD11b+, CD133+CD11b+, F4/80+, CD11b+, and F4/80+CD11b+) macrophages in bone marrow and blood compared with wild-type mice. Among the various CYP2C44-derived arachidonic acids, only 15-HETE decreased CD117+ cell numbers when applied to bone marrow cell cultures. Interestingly, CD133+ and von Willebrand factor-positive cells, which are derived from proangiogenic stem cells, are increased in the bone marrow, blood, and lungs of mice exposed to chronic hypoxia and in remodeled and occluded pulmonary arteries of CYP2C44-deficient mice. In conclusion, our results demonstrate that CYP2C44-derived 15-HETE plays a critical role in downregulating HSC proliferation and growth, because disruption of the Cyp2c44 gene increased HSCs that potentially contribute to chronic hypoxia-induced pulmonary arterial remodeling and occlusion.NEW & NOTEWORTHY This study demonstrates that cytochrome P-450 2C44 plays a critical role in controlling the phenotype of hematopoietic stem cells and that when this enzyme is knocked out, stem cells are differentiated. These stem cells give rise to increased circulating monocytes and macrophages and contribute to the pathogenesis of chronic hypoxia-induced pulmonary artery remodeling and hypertension.
Collapse
Affiliation(s)
- Ryota Hashimoto
- Department of Pharmacology, and Translation Cardiovascular Institute, School of Medicine, New York Medical College, Valhalla, New York.,Department of Physiology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Sachindra Raj Joshi
- Department of Pharmacology, and Translation Cardiovascular Institute, School of Medicine, New York Medical College, Valhalla, New York
| | - Houli Jiang
- Department of Pharmacology, and Translation Cardiovascular Institute, School of Medicine, New York Medical College, Valhalla, New York
| | - Jorge H Capdevila
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee; and
| | - Ivan F McMurtry
- Department of Pharmacology, University of South Alabama, Mobile, Alabama
| | - Michal Laniado Schwartzman
- Department of Pharmacology, and Translation Cardiovascular Institute, School of Medicine, New York Medical College, Valhalla, New York
| | - Sachin A Gupte
- Department of Pharmacology, and Translation Cardiovascular Institute, School of Medicine, New York Medical College, Valhalla, New York;
| |
Collapse
|