1
|
Zhang CL, Ma JJ, Li X, Yan HQ, Gui YK, Yan ZX, You MF, Zhang P. The role of transcytosis in the blood-retina barrier: from pathophysiological functions to drug delivery. Front Pharmacol 2025; 16:1565382. [PMID: 40308764 PMCID: PMC12040858 DOI: 10.3389/fphar.2025.1565382] [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: 01/24/2025] [Accepted: 03/31/2025] [Indexed: 05/02/2025] Open
Abstract
The blood-retina barrier (BRB) serves as a critical interface that separates the retina from the circulatory system, playing an essential role in preserving the homeostasis of the microenvironment within the retina. Specialized tight junctions and limited vesicle trafficking restrict paracellular and transcellular transport, respectively, thereby maintaining BRB barrier properties. Additionally, transcytosis of macromolecules through retinal vascular endothelial cells constitutes a primary mechanism for transporting substances from the vascular compartment into the surrounding tissue. This review summarizes the fundamental aspects of transcytosis including its function in the healthy retina, the biochemical properties of transcytosis, and the methodologies used to study this process. Furthermore, we discuss the current understanding of transcytosis in the context of pathological BRB breakdown and present recent findings that highlight significant advances in drug delivery to the retina based on transcytosis.
Collapse
Affiliation(s)
- Chun-Lin Zhang
- Department of Neurology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Jing-Jie Ma
- Department of Audit, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Xiang Li
- Department of Neurology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Hai-Qing Yan
- Department of Neurology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Yong-Kun Gui
- Department of Neurology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Zhi-Xin Yan
- Department of Neurology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Ming-Feng You
- Department of Neurology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Ping Zhang
- Department of Neurology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
2
|
Li C, Liu X, Liu J, Zhang X, Wu J, Ji X, Niu H, Xu Q. Chromatin accessibility and transcriptional landscape in PK-15 cells during early exposure to Aflatoxin B 1. Biochem Biophys Res Commun 2024; 731:150394. [PMID: 39024978 DOI: 10.1016/j.bbrc.2024.150394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 07/10/2024] [Accepted: 07/11/2024] [Indexed: 07/20/2024]
Abstract
Aflatoxin B1 (AFB1) not only causes significant losses in livestock production but also poses a serious threat to human health. It is the most carcinogenic among known chemicals. Pigs are more susceptible to AFB1 and experience a higher incidence. However, the molecular mechanism of the toxic effect of AFB1 remains unclear. In this study, we used assay for transposase-accessible chromatin using sequencing (ATAC-seq) and RNA-seq to uncover chromatin accessibility and gene expression dynamics in PK-15 cells during early exposure to AFB1. We observed that the toxic effects of AFB1 involve signaling pathways such as p53, PI3K-AKT, Hippo, MAPK, TLRs, apoptosis, autophagy, and cancer pathways. Basic leucine zipper (bZIP) transcription factors (TFs), including AP-1, Fos, JunB, and Fra2, play a crucial role in regulating the biological processes involved in AFB1 challenge. Several new TFs, such as BORIS, HNF1b, Atf1, and KNRNPH2, represent potential targets for the toxic mechanism of AFB1. In addition, it is crucial to focus on the concentration of intracellular zinc ions. These findings will contribute to a better understanding of the mechanisms underlying AFB1-induced nephrotoxicity and offer new molecular targets.
Collapse
Affiliation(s)
- Congcong Li
- College of Animal Science and Technology, Henan University of Animal Husbandry and Economy, Zhengzhou, Henan, China; Henan Pig Bio-breeding Research Institute, Zhengzhou, Henan, China; Henan Livestock and Poultry Genetic Resources Protection Engineering Technology Research Center, Zhengzhou, China.
| | - Xiangdong Liu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, China.
| | - Jiaxin Liu
- College of Animal Science and Technology, Henan University of Animal Husbandry and Economy, Zhengzhou, Henan, China.
| | - Xuanxuan Zhang
- College of Animal Science and Technology, Henan University of Animal Husbandry and Economy, Zhengzhou, Henan, China.
| | - Jiao Wu
- College of Animal Science and Technology, Henan University of Animal Husbandry and Economy, Zhengzhou, Henan, China.
| | - Xiangbo Ji
- College of Animal Science and Technology, Henan University of Animal Husbandry and Economy, Zhengzhou, Henan, China; Henan Key Laboratory of Unconventional Feed Resources Innovative Utilization, Henan University of Animal Husbandry and Economy, Zhengzhou, Henan, China.
| | - Hui Niu
- College of Animal Science and Technology, Henan University of Animal Husbandry and Economy, Zhengzhou, Henan, China.
| | - Qiuliang Xu
- College of Animal Science and Technology, Henan University of Animal Husbandry and Economy, Zhengzhou, Henan, China; Henan Pig Bio-breeding Research Institute, Zhengzhou, Henan, China; Henan Livestock and Poultry Genetic Resources Protection Engineering Technology Research Center, Zhengzhou, China.
| |
Collapse
|
3
|
Fan J, Zheng S, Wang M, Yuan X. The critical roles of caveolin-1 in lung diseases. Front Pharmacol 2024; 15:1417834. [PMID: 39380904 PMCID: PMC11458383 DOI: 10.3389/fphar.2024.1417834] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 09/09/2024] [Indexed: 10/10/2024] Open
Abstract
Caveolin-1 (Cav-1), a structural and functional component in the caveolae, plays a critical role in transcytosis, endocytosis, and signal transduction. Cav-1 has been implicated in the mediation of cellular processes by interacting with a variety of signaling molecules. Cav-1 is widely expressed in the endothelial cells, smooth muscle cells, and fibroblasts in the various organs, including the lungs. The Cav-1-mediated internalization and regulation of signaling molecules participate in the physiological and pathological processes. Particularly, the MAPK, NF-κB, TGFβ/Smad, and eNOS/NO signaling pathways have been involved in the regulatory effects of Cav-1 in lung diseases. The important effects of Cav-1 on the lungs indicate that Cav-1 can be a potential target for the treatment of lung diseases. A Cav-1 scaffolding domain peptide CSP7 targeting Cav-1 has been developed. In this article, we mainly discuss the structure of Cav-1 and its critical roles in lung diseases, such as pneumonia, acute lung injury (ALI), asthma, chronic obstructive pulmonary disease (COPD), pulmonary hypertension, pulmonary fibrosis, and lung cancer.
Collapse
Affiliation(s)
| | | | | | - Xiaoliang Yuan
- Department of Respiratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| |
Collapse
|
4
|
Azadi M, David AE. Enhancing Ocular Drug Delivery: The Effect of Physicochemical Properties of Nanoparticles on the Mechanism of Their Uptake by Human Cornea Epithelial Cells. ACS Biomater Sci Eng 2024; 10:429-441. [PMID: 38055935 DOI: 10.1021/acsbiomaterials.3c01144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/08/2023]
Abstract
This study investigates the effect of nanoparticle size and surface chemistry on interactions of the nanoparticles with human cornea epithelial cells (HCECs). Poly(lactic-co-glycolic) acid (PLGA) nanoparticles were synthesized using the emulsion-solvent evaporation method and surface modified with mucoadhesive (alginate [ALG] and chitosan [CHS]) and mucopenetrative (polyethylene glycol [PEG]) polymers. Particles were found to be monodisperse (polydispersity index (PDI) below 0.2), spherical, and with size and zeta potential ranging from 100 to 250 nm and from -25 to +15 mV, respectively. Evaluation of cytotoxicity with the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide (MTT) assay indicated that incubating cells with nanoparticles for 24 h at concentrations up to 100 μg/mL caused only mild toxicity (70-100% cell viability). Cellular uptake studies were conducted using an in vitro model developed with a monolayer of HCECs integrated with simulated mucosal solution. Evaluation of nanoparticle uptake revealed that energy-dependent endocytosis is the primary uptake mechanism. Among the different nanoparticles studied, 100 nm PLGA NPs and PEG-PLGA-150 NPs showed the highest levels of uptake by HCECs. Additionally, uptake studies in the presence of various inhibitors suggested that macropinocytosis and caveolae-mediated endocytosis are the dominant pathways. While clathrin-mediated endocytosis was found to also be partially responsible for nanoparticle uptake, phagocytosis did not play a role within the studied ranges of size and surface chemistries. These important findings could lead to improved nanoparticle-based formulations that could improve therapies for ocular diseases.
Collapse
Affiliation(s)
- Marjan Azadi
- Department of Chemical Engineering, Auburn University, Auburn, Alabama 36849, United States
| | - Allan E David
- Department of Chemical Engineering, Auburn University, Auburn, Alabama 36849, United States
| |
Collapse
|
5
|
Fayyaz AU, Sabbah MS, Dasari S, Griffiths LG, DuBrock HM, Wang Y, Charlesworth MC, Borlaug BA, Jenkins SM, Edwards WD, Redfield MM. Histologic and proteomic remodeling of the pulmonary veins and arteries in a porcine model of chronic pulmonary venous hypertension. Cardiovasc Res 2023; 119:268-282. [PMID: 35022664 PMCID: PMC10233294 DOI: 10.1093/cvr/cvac005] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 11/15/2021] [Accepted: 01/10/2022] [Indexed: 11/14/2022] Open
Abstract
AIMS In heart failure (HF), pulmonary venous hypertension (PVH) produces pulmonary hypertension (PH) with remodeling of pulmonary veins (PV) and arteries (PA). In a porcine PVH model, we performed proteomic-based bioinformatics to investigate unique pathophysiologic mechanisms mediating PA and PV remodeling. METHODS AND RESULTS Large PV were banded (PVH, n = 10) or not (Sham, n = 9) in piglets. At sacrifice, PV and PA were perfusion labelled for vessel-specific histology and proteomics. The PA and PV were separately sampled with laser-capture micro-dissection for mass spectrometry. Pulmonary vascular resistance [Wood Units; 8.6 (95% confidence interval: 6.3, 12.3) vs. 2.0 (1.7, 2.3)] and PA [19.9 (standard error of mean, 1.1) vs. 10.3 (1.1)] and PV [14.2 (1.2) vs. 7.6 (1.1)] wall thickness/external diameter (%) were increased in PVH (P < 0.05 for all). Similar numbers of proteins were identified in PA (2093) and PV (2085) with 94% overlap, but biological processes differed. There were more differentially expressed proteins (287 vs. 161), altered canonical pathways (17 vs. 3), and predicted upstream regulators (PUSR; 22 vs. 6) in PV than PA. In PA and PV, bioinformatics indicated activation of the integrated stress response and mammalian target of rapamycin signalling with dysregulated growth. In PV, there was also activation of Rho/Rho-kinase signalling with decreased actin cytoskeletal signalling and altered tight and adherens junctions, ephrin B, and caveolae-mediated endocytosis signalling; all indicating disrupted endothelial barrier function. Indeed, protein biomarkers and the top PUSR in PV (transforming growth factor-beta) suggested endothelial to mesenchymal transition in PV. Findings were similar in human autopsy specimens. CONCLUSION These findings provide new therapeutic targets to oppose pulmonary vascular remodeling in HF-related PH.
Collapse
Affiliation(s)
- Ahmed U Fayyaz
- Department of Cardiovascular Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
- Department of Laboratory Medicine and Pathology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Michael S Sabbah
- Department of Cardiovascular Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Surendra Dasari
- Division of Biomedical Statistics and Informatics, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Leigh G Griffiths
- Department of Cardiovascular Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Hilary M DuBrock
- Department of Cardiovascular Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Ying Wang
- Department of Cardiovascular Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - M Cristine Charlesworth
- Molecular Genome Facility Proteomics Core, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Barry A Borlaug
- Department of Cardiovascular Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Sarah M Jenkins
- Division of Biomedical Statistics and Informatics, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - William D Edwards
- Department of Laboratory Medicine and Pathology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Margaret M Redfield
- Department of Cardiovascular Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| |
Collapse
|
6
|
Mardi N, Haiaty S, Rahbarghazi R, Mobarak H, Milani M, Zarebkohan A, Nouri M. Exosomal transmission of viruses, a two-edged biological sword. Cell Commun Signal 2023; 21:19. [PMID: 36691072 PMCID: PMC9868521 DOI: 10.1186/s12964-022-01037-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Accepted: 12/28/2022] [Indexed: 01/24/2023] Open
Abstract
As a common belief, most viruses can egress from the host cells as single particles and transmit to uninfected cells. Emerging data have revealed en bloc viral transmission as lipid bilayer-cloaked particles via extracellular vesicles especially exosomes (Exo). The supporting membrane can be originated from multivesicular bodies during intra-luminal vesicle formation and autophagic response. Exo are nano-sized particles, ranging from 40-200 nm, with the ability to harbor several types of signaling molecules from donor to acceptor cells in a paracrine manner, resulting in the modulation of specific signaling reactions in target cells. The phenomenon of Exo biogenesis consists of multiple and complex biological steps with the participation of diverse constituents and molecular pathways. Due to similarities between Exo biogenesis and virus replication and the existence of shared pathways, it is thought that viruses can hijack the Exo biogenesis machinery to spread and evade immune cells. To this end, Exo can transmit complete virions (as single units or aggregates), separate viral components, and naked genetic materials. The current review article aims to scrutinize challenges and opportunities related to the exosomal delivery of viruses in terms of viral infections and public health. Video Abstract.
Collapse
Affiliation(s)
- Narges Mardi
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sanya Haiaty
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Imam Reza St., Golgasht St., Tabriz, Iran
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Halimeh Mobarak
- Stem Cell Research Center, Tabriz University of Medical Sciences, Imam Reza St., Golgasht St., Tabriz, Iran
| | - Morteza Milani
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Zarebkohan
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Nouri
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
7
|
Yang L, Tian J, Wang J, Zeng J, Wang T, Lin B, Linneman J, Li L, Niu Y, Gou D, Zhang Y. The protective role of EP300 in monocrotaline-induced pulmonary hypertension. Front Cardiovasc Med 2023; 10:1037217. [PMID: 36910531 PMCID: PMC9992637 DOI: 10.3389/fcvm.2023.1037217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 02/06/2023] [Indexed: 02/24/2023] Open
Abstract
Background Pulmonary hypertension (PH) is a lethal disease characterized by pulmonary vascular remodeling, which is mediated by the abnormal proliferation/migration of pulmonary arterial smooth muscle cells (PASMCs). Recent reports suggest the involvement of histone acetylation in PAH development and that histone deacetylase (HDAC) inhibitors have therapeutic potential for the treatment of PAH. EP300 is an acetyltransferase that plays diverse roles in cell proliferation, differentiation, and apoptosis. However, the functions of EP3000 in PH are rarely studied. Results In this work, we found that the expression of EP300 was increased in the pulmonary arteries of monocrotaline (MCT)-induced PH rats. Knockdown of EP300 by AAV-mediated shRNA exacerbated the PH, with a higher right ventricular systolic pressure (RVSP), right ventricular hypertrophy index (RVHI), and wall thickness in the pulmonary artery of MCT-induced PH rat. On the cellular level, the proliferation of PASMCs was promoted by EP300 knockdown. In addition, the expression of EP300 was increased in PASMCs by the overexpression of EGR1, while the deletion of EGR1 binding sites in the EP300 promoter region decreased the activity of EP300 promoter. Moreover, deleting the EP300 promoter region containing EGR1 binding sites using CRISPR/Cas9 abolished the upregulation of EP300 in MCT-induced rats and exacerbated MCT-induced PH. To summarize, our data indicate that EP300 upregulation mediated by EGR1 has a protective effect on MCT-induced PH. Conclusion These findings showed EP300 expression was increased in the MCT-induced PH model in rats, which could be mediated by EGR1; the EP300 also displayed the potential to provide protection from PH.
Collapse
Affiliation(s)
- Lei Yang
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Vascular Disease Research Center, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong, China
| | - Jinglin Tian
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Vascular Disease Research Center, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong, China
| | - Jun Wang
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Vascular Disease Research Center, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong, China
| | - Jie Zeng
- Department of Pulmonary and Critical Care Medicine, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Ting Wang
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Vascular Disease Research Center, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong, China
| | - Boya Lin
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Vascular Disease Research Center, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong, China
| | - John Linneman
- School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| | - Li Li
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Vascular Disease Research Center, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong, China
| | - Yanqin Niu
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Vascular Disease Research Center, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong, China
| | - Deming Gou
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Vascular Disease Research Center, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong, China
| | - Yunhui Zhang
- Department of Pulmonary and Critical Care Medicine, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| |
Collapse
|
8
|
Woo J, Clair GC, Williams SM, Feng S, Tsai CF, Moore RJ, Chrisler WB, Smith RD, Kelly RT, Paša-Tolić L, Ansong C, Zhu Y. Three-dimensional feature matching improves coverage for single-cell proteomics based on ion mobility filtering. Cell Syst 2022; 13:426-434.e4. [PMID: 35298923 PMCID: PMC9119937 DOI: 10.1016/j.cels.2022.02.003] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 12/04/2021] [Accepted: 02/17/2022] [Indexed: 12/13/2022]
Abstract
Single-cell proteomics (scProteomics) promises to advance our understanding of cell functions within complex biological systems. However, a major challenge of current methods is their inability to identify and provide accurate quantitative information for low-abundance proteins. Herein, we describe an ion-mobility-enhanced mass spectrometry acquisition and peptide identification method, transferring identification based on FAIMS filtering (TIFF), to improve the sensitivity and accuracy of label-free scProteomics. TIFF extends the ion accumulation times for peptide ions by filtering out singly charged ions. The peptide identities are assigned by a three-dimensional MS1 feature matching approach (retention time, accurate mass, and FAIMS compensation voltage). The TIFF method enabled unbiased proteome analysis to a depth of >1,700 proteins in single HeLa cells, with >1,100 proteins consistently identified. As a demonstration, we applied the TIFF method to obtain temporal proteome profiles of >150 single murine macrophage cells during lipopolysaccharide stimulation and identified time-dependent proteome changes. A record of this paper's transparent peer review process is included in the supplemental information.
Collapse
Affiliation(s)
- Jongmin Woo
- Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Geremy C Clair
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Sarah M Williams
- Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Song Feng
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Chia-Feng Tsai
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Ronald J Moore
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - William B Chrisler
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Richard D Smith
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Ryan T Kelly
- Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, WA 99354, USA; Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT 84602, USA
| | - Ljiljana Paša-Tolić
- Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Charles Ansong
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Ying Zhu
- Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, WA 99354, USA.
| |
Collapse
|
9
|
Abstract
The development of pulmonary hypertension (PH) is common and has adverse prognostic implications in patients with heart failure due to left heart disease (LHD), and thus far, there are no known treatments specifically for PH-LHD, also known as group 2 PH. Diagnostic thresholds for PH-LHD, and clinical classification of PH-LHD phenotypes, continue to evolve and, therefore, present a challenge for basic and translational scientists actively investigating PH-LHD in the preclinical setting. Furthermore, the pathobiology of PH-LHD is not well understood, although pulmonary vascular remodeling is thought to result from (1) increased wall stress due to increased left atrial pressures; (2) hemodynamic congestion-induced decreased shear stress in the pulmonary vascular bed; (3) comorbidity-induced endothelial dysfunction with direct injury to the pulmonary microvasculature; and (4) superimposed pulmonary arterial hypertension risk factors. To ultimately be able to modify disease, either by prevention or treatment, a better understanding of the various drivers of PH-LHD, including endothelial dysfunction, abnormalities in vascular tone, platelet aggregation, inflammation, adipocytokines, and systemic complications (including splanchnic congestion and lymphatic dysfunction) must be further investigated. Here, we review the diagnostic criteria and various hemodynamic phenotypes of PH-LHD, the potential biological mechanisms underlying this disorder, and pressing questions yet to be answered about the pathobiology of PH-LHD.
Collapse
Affiliation(s)
- Jessica H Huston
- Division of Cardiology, Department of Internal Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA (J.H.H.)
| | - Sanjiv J Shah
- Division of Cardiology, Department of Medicine, Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL (S.J.S.)
| |
Collapse
|
10
|
Nagano H, Ogata S, Ito S, Masuda T, Ohtsuki S. Knockdown of podocalyxin post-transcriptionally induces the expression and activity of ABCB1/MDR1 in human brain microvascular endothelial cells. J Pharm Sci 2022; 111:1812-1819. [PMID: 35182544 DOI: 10.1016/j.xphs.2022.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 02/11/2022] [Accepted: 02/12/2022] [Indexed: 10/19/2022]
Abstract
Podocalyxin (PODXL) is a highly sialylated transmembrane protein that is expressed on the luminal membrane of brain microvascular endothelial cells. To clarify the role of PODXL in the blood-brain barrier (BBB), the present study aimed to investigate the effect of PODXL-knockdown on protein expression, especially the expression of ABCB1/MDR1, in human microvascular endothelial cells (hCMEC/D3). By quantitative proteomics, gene ontology enrichment with differentially expressed proteins showed that PODXL-knockdown influenced the immune response and intracellular trafficking. Among transporters, the protein expression of ABCB1/MDR1 and ABCG2/BCRP was significantly elevated by approximately 2-fold in the PODXL-knockdown cells. In the knockdown cells, the efflux activity of ABCB1/MDR1 was significantly increased, while its mRNA expression was not significantly different from that of the control cells. As receptors and tight junction proteins, levels of low-density lipoprotein receptor-related protein 1 and occludin were significantly increased, while those of transferrin receptor and claudin-11 were significantly decreased in the knockdown cells. The present results suggest that PODXL functions as a modulator of BBB function, including transport, tight junctions, and immune responses. Furthermore, PODXL post-transcriptionally regulates the protein expression and efflux activity of ABCB1/MDR1 at the BBB, which may affect drug distribution in the brain.
Collapse
Key Words
- Blood-brain barrier, brain microvascular endothelial cells, ABCB1, MDR1, podocalyxin, proteomics, regulation, List of Abbreviations, BMECs
- Bood-brain barrier, HFD
- Brain microvascular endothelial cells, BBB
- Control hCMEC/D3 cells, shPODXL
- High-fat diet, LRP1
- Low-density lipoprotein receptor-related protein 1, MS
- Mass spectrometry, PODXL
- PODXL-knockdown hCMEC/D3 cells, SEM
- Podocalyxin, shNT
- Standard error of the mean, TFRC
- Transferrin receptor
Collapse
Affiliation(s)
- Hinako Nagano
- Department of Pharmaceutical Microbiology, School of Pharmacy, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan
| | - Seiryo Ogata
- Department of Pharmaceutical Microbiology, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan
| | - Shingo Ito
- Department of Pharmaceutical Microbiology, School of Pharmacy, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan; Department of Pharmaceutical Microbiology, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan; Department of Pharmaceutical Microbiology, Faculty of Life Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan
| | - Takeshi Masuda
- Department of Pharmaceutical Microbiology, School of Pharmacy, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan; Department of Pharmaceutical Microbiology, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan; Department of Pharmaceutical Microbiology, Faculty of Life Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan
| | - Sumio Ohtsuki
- Department of Pharmaceutical Microbiology, School of Pharmacy, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan; Department of Pharmaceutical Microbiology, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan; Department of Pharmaceutical Microbiology, Faculty of Life Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan.
| |
Collapse
|
11
|
Li C, Huang J, Tang H, Liu B, Zhou X. Revealing Cavin-2 Gene Function in Lung Based on Multi-Omics Data Analysis Method. Front Cell Dev Biol 2022; 9:827108. [PMID: 35174175 PMCID: PMC8841408 DOI: 10.3389/fcell.2021.827108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 12/15/2021] [Indexed: 11/23/2022] Open
Abstract
Research points out that it is particularly important to comprehensively evaluate immune microenvironmental indicators and gene mutation characteristics to select the best treatment plan. Therefore, exploring the relevant genes of pulmonary injury is an important basis for the improvement of survival. In recent years, with the massive production of omics data, a large number of computational methods have been applied in the field of biomedicine. Most of these computational methods are devel-oped for a certain type of diseases or whole diseases. Algorithms that specifically identify genes associated with pulmonary injury have not yet been developed. To fill this gap, we developed a novel method, named AdaRVM, to identify pulmonary injury-related genes in large scale. AdaRVM is the fusion of Adaboost and Relevance Vector Machine (RVM) to achieve fast and high-precision pattern recognition of pulmonary injury genetic mechanism. AdaRVM found that Cavin-2 gene has strong potential to be related to pulmonary injury. As we known, the formation and function of Caveolae are mediated by two family proteins: Caveolin and Cavin. Many studies have explored the role of Caveolin proteins, but people still knew little about Cavin family members. To verify our method and reveal the functions of cavin-2, we integrated six genome-wide association studies (GWAS) data related to lung function traits, four expression Quantitative Trait Loci (eQTL) data, and one methylation Quantitative Trait Loci (mQTL) data by Summary data level Mendelian Randomization (SMR). We found strong relationship between cavin-2 and canonical signaling pathways ERK1/2, AKT, and STAT3 which are all known to be related to lung injury.
Collapse
Affiliation(s)
- Changsheng Li
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jingyu Huang
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Hexiao Tang
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Bing Liu
- Department of Pulmonary and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
- Wuhan Research Center for Infectious Diseases and Cancer, Chinese Academy of Medical Sciences, Wuhan, China
| | - Xuefeng Zhou
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
- *Correspondence: Xuefeng Zhou,
| |
Collapse
|
12
|
Liu J, Ke X, Wang L, Zhang Y, Yang J. Deficiency of cold-inducible RNA-binding protein exacerbated monocrotaline-induced pulmonary artery hypertension through Caveolin1 and CAVIN1. J Cell Mol Med 2021; 25:4732-4743. [PMID: 33755319 PMCID: PMC8107102 DOI: 10.1111/jcmm.16437] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 02/16/2021] [Accepted: 02/18/2021] [Indexed: 12/18/2022] Open
Abstract
Cold‐inducible RNA‐binding protein (CIRP) was a crucial regulator in multiple diseases. However, its role in pulmonary artery hypertension (PAH) is still unknown. Here, we first established monocrotaline (MCT)‐induced rat PAH model and discovered that CIRP was down‐regulated predominantly in the endothelium of pulmonary artery after MCT injection. We then generated Cirp‐knockout (Cirp‐KO) rats, which manifested severer PAH with exacerbated endothelium damage in response to MCT. Subsequently, Caveolin1 (Cav1) and Cavin1 were identified as downstream targets of CIRP in MCT‐induced PAH, and the decreased expression of these two genes aggravated the injury and apoptosis of pulmonary artery endothelium. Moreover, CIRP deficiency intensified monocrotaline pyrrole (MCTP)‐induced rat pulmonary artery endothelial cells (rPAECs) injuries both in vivo and in vitro, which was counteracted by Cav1 or Cavin1 overexpression. In addition, CIRP regulated the proliferative effect of conditioned media from MCTP‐treated rPAECs on rat pulmonary artery smooth muscle cells, which partially explained the exceedingly thickened pulmonary artery intimal media in Cirp‐KO rats after MCT treatment. These results demonstrated that CIRP acts as a critical protective factor in MCT‐induced rat PAH by directly regulating CAV1 and CAVIN1 expression, which may facilitate the development of new therapeutic targets for the intervention of PAH.
Collapse
Affiliation(s)
- Jingjing Liu
- Key Laboratory of Arrhythmias, Ministry of Education, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China.,Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China.,Institute of Medical Genetics, Tongji University School of Medicine, Shanghai, China
| | - Xianting Ke
- Key Laboratory of Arrhythmias, Ministry of Education, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China.,Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China.,Institute of Medical Genetics, Tongji University School of Medicine, Shanghai, China
| | - Luxin Wang
- Key Laboratory of Arrhythmias, Ministry of Education, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China.,Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China.,Institute of Medical Genetics, Tongji University School of Medicine, Shanghai, China
| | - Yangyang Zhang
- Key Laboratory of Arrhythmias, Ministry of Education, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China.,Department of Cardiovascular Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jian Yang
- Key Laboratory of Arrhythmias, Ministry of Education, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China.,Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China.,Institute of Medical Genetics, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
13
|
Madni A, Rehman S, Sultan H, Khan MM, Ahmad F, Raza MR, Rai N, Parveen F. Mechanistic Approaches of Internalization, Subcellular Trafficking, and Cytotoxicity of Nanoparticles for Targeting the Small Intestine. AAPS PharmSciTech 2020; 22:3. [PMID: 33221968 PMCID: PMC7680634 DOI: 10.1208/s12249-020-01873-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 11/05/2020] [Indexed: 12/12/2022] Open
Abstract
Targeting the small intestine employing nanotechnology has proved to be a more effective way for site-specific drug delivery. The drug targeting to the small intestine can be achieved via nanoparticles for its optimum bioavailability within the systemic circulation. The small intestine is a remarkable candidate for localized drug delivery. The intestine has its unique properties. It has a less harsh environment than the stomach, provides comparatively more retention time, and possesses a greater surface area than other parts of the gastrointestinal tract. This review focuses on elaborating the intestinal barriers and approaches to overcome these barriers for internalizing nanoparticles and adopting different cellular trafficking pathways. We have discussed various factors that contribute to nanocarriers' cellular uptake, including their surface chemistry, surface morphology, and functionalization of nanoparticles. Furthermore, the fate of nanoparticles after their uptake at cellular and subcellular levels is also briefly explained. Finally, we have delineated the strategies that are adopted to determine the cytotoxicity of nanoparticles.
Collapse
Affiliation(s)
- Asadullah Madni
- Department of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur, Pakistan.
| | - Sadia Rehman
- Department of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | - Humaira Sultan
- Department of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | | | - Faiz Ahmad
- Departments of Mechanical Engineering, Universiti Teknologi PETRONAS, Seri Iskandar, Malaysia
| | - M Rafi Raza
- Department of Mechanical Engineering, COMSATS University Islamabad, Sahiwal Campus, Sahiwal, Pakistan
| | - Nadia Rai
- Department of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | - Farzana Parveen
- Department of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| |
Collapse
|
14
|
Mutgan AC, Jandl K, Kwapiszewska G. Endothelial Basement Membrane Components and Their Products, Matrikines: Active Drivers of Pulmonary Hypertension? Cells 2020; 9:cells9092029. [PMID: 32899187 PMCID: PMC7563239 DOI: 10.3390/cells9092029] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/27/2020] [Accepted: 08/29/2020] [Indexed: 12/19/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a vascular disease that is characterized by elevated pulmonary arterial pressure (PAP) due to progressive vascular remodeling. Extracellular matrix (ECM) deposition in pulmonary arteries (PA) is one of the key features of vascular remodeling. Emerging evidence indicates that the basement membrane (BM), a specialized cluster of ECM proteins underlying the endothelium, may be actively involved in the progression of vascular remodeling. The BM and its steady turnover are pivotal for maintaining appropriate vascular functions. However, the pathologically elevated turnover of BM components leads to an increased release of biologically active short fragments, which are called matrikines. Both BM components and their matrikines can interfere with pivotal biological processes, such as survival, proliferation, adhesion, and migration and thus may actively contribute to endothelial dysfunction. Therefore, in this review, we summarize the emerging role of the BM and its matrikines on the vascular endothelium and further discuss its implications on lung vascular remodeling in pulmonary hypertension.
Collapse
Affiliation(s)
- Ayse Ceren Mutgan
- Otto Loewi Research Center, Division of Physiology, Medical University of Graz, 8010 Graz, Austria;
| | - Katharina Jandl
- Ludwig Boltzmann Institute for Lung Vascular Research, 8010 Graz, Austria;
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, 8010 Graz, Austria
| | - Grazyna Kwapiszewska
- Otto Loewi Research Center, Division of Physiology, Medical University of Graz, 8010 Graz, Austria;
- Ludwig Boltzmann Institute for Lung Vascular Research, 8010 Graz, Austria;
- Correspondence:
| |
Collapse
|
15
|
Filippini A, D’Alessio A. Caveolae and Lipid Rafts in Endothelium: Valuable Organelles for Multiple Functions. Biomolecules 2020; 10:biom10091218. [PMID: 32825713 PMCID: PMC7563503 DOI: 10.3390/biom10091218] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 08/17/2020] [Accepted: 08/18/2020] [Indexed: 12/22/2022] Open
Abstract
Caveolae are flask-shaped invaginations of the plasma membrane found in numerous cell types and are particularly abundant in endothelial cells and adipocytes. The lipid composition of caveolae largely matches that of lipid rafts microdomains that are particularly enriched in cholesterol, sphingomyelin, glycosphingolipids, and saturated fatty acids. Unlike lipid rafts, whose existence remains quite elusive in living cells, caveolae can be clearly distinguished by electron microscope. Despite their similar composition and the sharing of some functions, lipid rafts appear more heterogeneous in terms of size and are more dynamic than caveolae. Following the discovery of caveolin-1, the first molecular marker as well as the unique scaffolding protein of caveolae, we have witnessed a remarkable increase in studies aimed at investigating the role of these organelles in cell functions and human disease. The goal of this review is to discuss the most recent studies related to the role of caveolae and caveolins in endothelial cells. We first recapitulate the major embryological processes leading to the formation of the vascular tree. We next discuss the contribution of caveolins and cavins to membrane biogenesis and cell response to extracellular stimuli. We also address how caveolae and caveolins control endothelial cell metabolism, a central mechanism involved in migration proliferation and angiogenesis. Finally, as regards the emergency caused by COVID-19, we propose to study the caveolar platform as a potential target to block virus entry into endothelial cells.
Collapse
Affiliation(s)
- Antonio Filippini
- Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Unit of Histology and Medical Embryology, Sapienza University of Rome, 00161 Roma, Italy;
| | - Alessio D’Alessio
- Dipartimento di Scienze della Vita e Sanità Pubblica, Sezione di Istologia ed Embriologia, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario “Agostino Gemelli”, IRCCS, 00168 Roma, Italia
- Correspondence:
| |
Collapse
|
16
|
Abstract
Transcytosis of macromolecules through lung endothelial cells is the primary route of transport from the vascular compartment into the interstitial space. Endothelial transcytosis is mostly a caveolae-dependent process that combines receptor-mediated endocytosis, vesicle trafficking via actin-cytoskeletal remodeling, and SNARE protein directed vesicle fusion and exocytosis. Herein, we review the current literature on caveolae-mediated endocytosis, the role of actin cytoskeleton in caveolae stabilization at the plasma membrane, actin remodeling during vesicle trafficking, and exocytosis of caveolar vesicles. Next, we provide a concise summary of experimental methods employed to assess transcytosis. Finally, we review evidence that transcytosis contributes to the pathogenesis of acute lung injury. © 2020 American Physiological Society. Compr Physiol 10:491-508, 2020.
Collapse
Affiliation(s)
- Joshua H. Jones
- Department of Pharmacology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Richard D. Minshall
- Department of Pharmacology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA,Department of Anesthesiology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA,Correspondence to
| |
Collapse
|
17
|
Impact of nitrate therapy on the expression of caveolin-1 and its phosphorylated isoform in lungs in the model of monocrotaline induced pulmonary hypertension. EUROPEAN PHARMACEUTICAL JOURNAL 2018. [DOI: 10.2478/afpuc-2018-0002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Aim: Nitric oxide signalling pathway showed to be one of the crucial factors in the treatment and pathogenesis of pulmonary arterial hypertension. The aim of this study was to determine the effect of administration of inorganic nitrate, NaNO3, on the expression of caveolin-1 and its phosphorylated isoform (pTyr14Cav-1) in lungs in the experimental model of monocrotaline induced pulmonary hypertension.
Methods: 10 weeks old male Wistar rats were subcutaneously injected with 60 mg/kg dose of monocrotaline (MCT) or vehicle (CON). Twelve days after the injection, part of the MCT group was receiving 0.3 mM NaNO3 (MCT+N0.3) daily in the drinking water and rest was receiving 0.08% NaCl solution. Four weeks after MCT administration, the rats were sacrificed in CO2. Protein expression in lungs was determined by western blot.
Results: We observed a significant decrease in the caveolin-1 expression and a significant shift towards the expression of pTyr14Cav-1 in the group treated with nitrate (p < 0.05).
Conclusion: NaNO3 administration affected the expression of caveolin-1 and the ratio of its active (phosphorylated) isoform increased.
Collapse
|
18
|
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: 0.9] [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
|