1
|
Hsu JCN, Chiu KT, Chen CH, Wang CH, Shyue SK, Lee TS. HMGB1 Regulates Adipocyte Lipolysis via Caveolin-1 Signaling: Implications for Metabolic and Cardiovascular Diseases. Int J Mol Sci 2025; 26:4222. [PMID: 40362460 PMCID: PMC12071352 DOI: 10.3390/ijms26094222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2025] [Revised: 04/24/2025] [Accepted: 04/26/2025] [Indexed: 05/15/2025] Open
Abstract
High-mobility group box 1 (HMGB1) is a nuclear protein that can be secreted or released into the extracellular environment during cellular stress, functioning as a damage-associated molecular pattern molecule. This study investigates the role of HMGB1 in adipocyte development and metabolism, explicitly examining its interaction with β3-adrenergic receptor-mediated lipolysis and caveolin-1 (CAV1) regulation, which may influence cardiovascular risk factors. Using 3T3-L1 preadipocytes and mouse embryonic fibroblasts, we demonstrated that HMGB1 expression increases progressively during adipogenesis, reaching peak levels in mature adipocytes. While exogenous HMGB1 treatment did not affect preadipocyte proliferation or differentiation, it inhibited lipolysis in mature adipocytes. Mechanistically, HMGB1 suppressed β3-adrenergic receptor agonist CL-316,243-induced hormone-sensitive lipase activation by reducing protein kinase A-mediated phosphorylation and attenuating extracellular signal-regulated kinase signaling without affecting upstream cyclic AMP levels. We discovered a novel regulatory mechanism wherein CAV1 physically interacts with HMGB1 in mature adipocytes, with c-Src-dependent CAV1 phosphorylation functioning as a negative regulator of HMGB1 secretion. This finding was confirmed in CAV1-deficient models, which displayed increased HMGB1 secretion and diminished lipolytic activity both in vitro and in vivo. Furthermore, administering HMGB1-neutralizing antibodies to wild-type mice enhanced fasting-induced lipolysis, establishing circulating HMGB1 as a crucial antilipolytic factor. These findings reveal HMGB1's previously uncharacterized role in adipose tissue metabolism as a negative regulator of lipolysis through CAV1-dependent mechanisms. This work provides new insights into adipose tissue metabolism regulation and identifies potential therapeutic targets for obesity-related metabolic disorders and cardiovascular diseases.
Collapse
Affiliation(s)
- Julia Chu-Ning Hsu
- Department of Veterinary Medicine, College of Veterinary Medicine, National Chung Hsing University, 145, Xingda Road, South District, Taichung 402202, Taiwan;
| | - Kuan-Ting Chiu
- Department of Physiology, School of Medicine, National Yang-Ming University, 155, Sec. 2, Linong Street, Beitou District, Taipei 112304, Taiwan
| | - Chia-Hui Chen
- Graduate Institute and Department of Physiology, College of Medicine, National Taiwan University, 1, Sec. 1, Jenai Road, Zhongzheng District, Taipei 100233, Taiwan
| | - Chih-Hsien Wang
- Cardiovascular Surgery, Department of Surgery, National Taiwan University Hospital and College of Medicine, 7, Chungshan South Road, Zhongzheng District, Taipei 100225, Taiwan
| | - Song-Kun Shyue
- Institute of Biomedical Sciences, Academia Sinica, 128, Sec. 2, Academia Road, Nankang District, Taipei 115201, Taiwan
| | - Tzong-Shyuan Lee
- Graduate Institute and Department of Physiology, College of Medicine, National Taiwan University, 1, Sec. 1, Jenai Road, Zhongzheng District, Taipei 100233, Taiwan
| |
Collapse
|
2
|
Zhu J, Lin Q, Zhu H, Xie S, Nie S. Toxicity mechanism analysis of cGAS-STING-TBK1 signaling pathway small molecule modulator based on network toxicology and molecular docking strategy: quinacrine acetate as an example. Front Chem 2025; 13:1584588. [PMID: 40331038 PMCID: PMC12052562 DOI: 10.3389/fchem.2025.1584588] [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: 02/27/2025] [Accepted: 03/25/2025] [Indexed: 05/08/2025] Open
Abstract
Objective This study aims to investigate the toxicity characteristics and mechanisms of quinacrine acetate, a small molecule modulator of the cGAS-STING-TBK1 signaling pathway, and to establish and validate the application value of network toxicology analysis strategy. Methods ProTox and ADMETlab platforms were used to evaluate the toxic effects of quinacrine acetate on human tissues and organs. Potential targets associated with quinacrine acetate toxicity were identified through ChEMBL, STITCH, GeneCards, OMIM, and TD databases. GO and KEGG analyses were employed to elucidate related functions and molecular mechanisms. STRING and Cytoscape software were utilized to identify key hub genes, while molecular docking validation was performed using the CB-Dock2 database. Based on toxicity analysis results, COPD was selected as a disease model, and GEO database was used to analyze the expression characteristics, immune correlation, and drug target value of hub genes in COPD. Results ProTox and ADMETlab analyses revealed that quinacrine acetate exhibited significant toxicity to the respiratory system (toxicity level 4, risk coefficient 0.959). Through integrated multi-database analysis, 14 potential targets related to quinacrine acetate-induced respiratory system toxicity were identified. GO and KEGG pathway analyses indicated that quinacrine acetate-induced respiratory toxicity was primarily mediated through metabolic pathways. Network analysis via STRING and Cytoscape identified AKT1, PLA2G4A, and ALOX5 as three core targets. Molecular docking results confirmed strong binding affinity between quinacrine acetate and these core targets. In COPD patients, PLA2G4A and ALOX5 showed significantly upregulated expression, with hub gene ROC curve AUC value reaching 0.829, demonstrating good diagnostic value. Further immune correlation analysis revealed that ALOX5 and PLA2G4A were closely associated with various immune cell expressions and served as targets for multiple drugs including histamine, melittin, and formic acid. Conclusion This study demonstrates that quinacrine acetate may influence the progression and risk of respiratory system diseases by regulating metabolic pathways. The findings provide not only a theoretical foundation for understanding the molecular mechanisms of quinacrine acetate-induced respiratory toxicity but also new perspectives and methodological references for evaluating the toxic effects of small molecule compounds in respiratory diseases. Therefore, we demonstrates the practical application value of network toxicology as an efficient predictive tool for identifying potential toxicity targets and pathways, which can guide subsequent experimental validation and provide mechanistic insights that traditional toxicology approaches might miss.
Collapse
Affiliation(s)
- Jinchao Zhu
- School of Health Science and Engineering University of Shanghai for Science and Technology, Shanghai, China
| | - Qingyuan Lin
- Department of Pathology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Honglin Zhu
- School of Health Science and Engineering University of Shanghai for Science and Technology, Shanghai, China
| | - Siqi Xie
- Sichuan Cancer Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Shengdong Nie
- School of Health Science and Engineering University of Shanghai for Science and Technology, Shanghai, China
| |
Collapse
|
3
|
Li Z, Dang Q, Liu C, Liu Y, Wang C, Zhao F, Wang Q, Min W. Caveolin Regulates the Transport Mechanism of the Walnut-Derived Peptide EVSGPGYSPN to Penetrate the Blood-Brain Barrier. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:19786-19799. [PMID: 39187786 DOI: 10.1021/acs.jafc.4c03291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/28/2024]
Abstract
Bioactive peptides, derived from short protein fragments, are recognized for their neuroprotective properties and potential therapeutic applications in treating central nervous system (CNS) diseases. However, a significant challenge for these peptides is their ability to penetrate the blood-brain barrier (BBB). EVSGPGYSPN (EV-10) peptide, a walnut-derived peptide, has demonstrated promising neuroprotective effects in vivo. This study aimed to investigate the transportability of EV-10 across the BBB, explore its capacity to penetrate this barrier, and elucidate the regulatory mechanisms underlying peptide-induced cellular internalization and transport pathways within the BBB. The results indicated that at a concentration of 100 μM and osmotic time of 4 h, the apparent permeability coefficient of EV-10 was Papp = 8.52166 ± 0.58 × 10-6 cm/s. The penetration efficiency of EV-10 was influenced by time, concentration, and temperature. Utilizing Western blot analysis, immunofluorescence, and flow cytometry, in conjunction with the caveolin (Cav)-specific inhibitor M-β-CD, we confirmed that EV-10 undergoes transcellular transport through a Cav-dependent endocytosis pathway. Notably, the tight junction proteins ZO-1, occludin, and claudin-5 were not disrupted by EV-10. Throughout its transport, EV-10 was localized within the mitochondria, Golgi apparatus, endoplasmic reticulum, lysosomes, endosomes, and cell membranes. Moreover, Cav-1 overexpression facilitated the release of EV-10 from lysosomes. Evidence of EV-10 accumulation was observed in mouse brains using brain slice scans. This study is the first to demonstrate that Cav-1 can facilitate the targeted delivery of walnut-derived peptide to the brain, laying a foundation for the development of functional foods aimed at CNS disease intervention.
Collapse
Affiliation(s)
- Zehui Li
- College of Food Science and Engineering, Jilin Agricultural University, ChangChun, Jilin 130118, P. R. China
- State Key Laboratory of Subtropical Silviculture, Zhejiang A&F University, Hangzhou 311300, P. R. China
- College of Food and Health, Zhejiang A&F University, Hangzhou, Zhejiang 311300, P. R. China
| | - Qiao Dang
- College of Food Science and Engineering, Jilin Agricultural University, ChangChun, Jilin 130118, P. R. China
| | - Chunlei Liu
- College of Food Science and Engineering, Jilin Agricultural University, ChangChun, Jilin 130118, P. R. China
| | - Yan Liu
- State Key Laboratory of Subtropical Silviculture, Zhejiang A&F University, Hangzhou 311300, P. R. China
- College of Food and Health, Zhejiang A&F University, Hangzhou, Zhejiang 311300, P. R. China
| | - Chongchong Wang
- State Key Laboratory of Subtropical Silviculture, Zhejiang A&F University, Hangzhou 311300, P. R. China
- College of Food and Health, Zhejiang A&F University, Hangzhou, Zhejiang 311300, P. R. China
| | - Fanrui Zhao
- State Key Laboratory of Subtropical Silviculture, Zhejiang A&F University, Hangzhou 311300, P. R. China
- College of Food and Health, Zhejiang A&F University, Hangzhou, Zhejiang 311300, P. R. China
| | - Qianqian Wang
- State Key Laboratory of Subtropical Silviculture, Zhejiang A&F University, Hangzhou 311300, P. R. China
- College of Food and Health, Zhejiang A&F University, Hangzhou, Zhejiang 311300, P. R. China
| | - Weihong Min
- State Key Laboratory of Subtropical Silviculture, Zhejiang A&F University, Hangzhou 311300, P. R. China
- College of Food and Health, Zhejiang A&F University, Hangzhou, Zhejiang 311300, P. R. China
| |
Collapse
|
4
|
Mejlachowicz D, Lassiaz P, Zola M, Leclercq B, Gélizé E, Achiedo S, Zhao M, Rousseau A, Behar-Cohen F. Identification of Structures Labeled by Indocyanine Green in the Rat Choroid and Retina Can Guide Interpretation of Indocyanine Green Angiography. Invest Ophthalmol Vis Sci 2024; 65:25. [PMID: 38193758 PMCID: PMC10784846 DOI: 10.1167/iovs.65.1.25] [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: 08/13/2023] [Accepted: 12/07/2023] [Indexed: 01/10/2024] Open
Abstract
Purpose Indocyanine green (ICG) is an albumin and lipoprotein binding dye absorbing in the far red used in angiography to visualize choroidal vessels (ICG angiography [ICGA]). To guide interpretation, ICG transport in the choroid, RPE, and retina of rats was studied. Methods Two conditions were used: RPE/choroid organoculture, incubated for 45 minutes in DMEM medium, 1% fetal bovine serum containing 0.25 mg/mL ICG and RPE/choroid and neural retina flat-mounts at 1 and 6 hours after intravenous ICG injection. Early and late sequences of ICGA were recorded until 6 hours. Ultra-deep red confocal microscope was used to localize ICG in flat-mounts and immunohistochemistry was performed for caveolin-1, tryptase (mast cell marker), and tubulin β3 (a nerve marker). Results In the organoculture, ICG penetrated homogeneously in the cytoplasm and stained the membranes of the RPE. At 1 hour after intravenous injection, ICG appeared in fine granules in RPE, partly labeled with caveolin-1 and decreasing at 6 hours. At 1 hour and 6 hours, ICG was found in the retinal vessels, faintly in the inner retina, and in the photoreceptor outer segments at 6 hours. In the choroid, ICG colocalized with mast cells, immunostained with tryptase, and accumulated along the large tubulin β3-labeled nerve bundles. The hypothesis was raised on the interpretation of late ICGA infrared photography in case of transthyretin amyloidosis with neuropathy. Conclusions Beside being a vascular dye, ICG is transported from the vessels to the RPE toward the outer retina. It stains mast cells and large choroidal nerves. These observations could help the analysis of ICGA images.
Collapse
Affiliation(s)
- Dan Mejlachowicz
- Centre de Recherche des Cordeliers, INSERM, Université Paris Cité, Sorbonne Université, Physiopathology of Ocular Diseases: Therapeutic Innovations, Paris, France
| | - Patricia Lassiaz
- Centre de Recherche des Cordeliers, INSERM, Université Paris Cité, Sorbonne Université, Physiopathology of Ocular Diseases: Therapeutic Innovations, Paris, France
| | - Marta Zola
- Centre de Recherche des Cordeliers, INSERM, Université Paris Cité, Sorbonne Université, Physiopathology of Ocular Diseases: Therapeutic Innovations, Paris, France
- Department of Ophthalmology, Hopital Foch, Suresnes, France
| | - Bastien Leclercq
- Centre de Recherche des Cordeliers, INSERM, Université Paris Cité, Sorbonne Université, Physiopathology of Ocular Diseases: Therapeutic Innovations, Paris, France
| | - Emmanuelle Gélizé
- Centre de Recherche des Cordeliers, INSERM, Université Paris Cité, Sorbonne Université, Physiopathology of Ocular Diseases: Therapeutic Innovations, Paris, France
| | - Seiki Achiedo
- Centre de Recherche des Cordeliers, INSERM, Université Paris Cité, Sorbonne Université, Physiopathology of Ocular Diseases: Therapeutic Innovations, Paris, France
| | - Min Zhao
- Centre de Recherche des Cordeliers, INSERM, Université Paris Cité, Sorbonne Université, Physiopathology of Ocular Diseases: Therapeutic Innovations, Paris, France
| | - Antoine Rousseau
- Department of Ophthalmology, Bicêtre Hospital, Assistance Publique - Hôpitaux de Paris, Paris-Saclay University, French Reference Center for hereditary transthyretin amyloidosis (NNERF), French Reference Network for rare Ophthalmic diseases (OPHTARA), Le Kremlin-Bicêtre, France
| | - Francine Behar-Cohen
- Centre de Recherche des Cordeliers, INSERM, Université Paris Cité, Sorbonne Université, Physiopathology of Ocular Diseases: Therapeutic Innovations, Paris, France
- Ophthalmopole Cochin University Hospital, Assistance Publique-Hôpitaux de Paris, France
- Department of Ophthalmology, Hopital Foch, Suresnes, France
| |
Collapse
|
5
|
Gan C, Li W, Xu J, Pang L, Tang L, Yu S, Li A, Ge H, Huang R, Cheng H. Advances in the study of the molecular biological mechanisms of radiation-induced brain injury. Am J Cancer Res 2023; 13:3275-3299. [PMID: 37693137 PMCID: PMC10492106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Accepted: 07/12/2023] [Indexed: 09/12/2023] Open
Abstract
Radiation therapy is one of the most commonly used treatments for head and neck cancers, but it often leads to radiation-induced brain injury. Patients with radiation-induced brain injury have a poorer quality of life, and no effective treatments are available. The pathogenesis of this condition is unknown. This review summarizes the molecular biological mechanism of radiation-induced brain injury and provides research directions for future studies. The molecular mechanisms of radiation-induced brain injury are diverse and complex. Radiation-induced chronic neuroinflammation, destruction of the blood-brain barrier, oxidative stress, neuronal damage, and physiopathological responses caused by specific exosome secretion lead to radiation-induced brain injury.
Collapse
Affiliation(s)
- Chen Gan
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical UniversityHefei, Anhui, China
- Department of Oncology, Anhui Medical UniversityHefei, Anhui, China
| | - Wen Li
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical UniversityHefei, Anhui, China
- Department of Oncology, Anhui Medical UniversityHefei, Anhui, China
| | - Jian Xu
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical UniversityHefei, Anhui, China
- Department of Oncology, Anhui Medical UniversityHefei, Anhui, China
| | - Lulian Pang
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical UniversityHefei, Anhui, China
- Department of Oncology, Anhui Medical UniversityHefei, Anhui, China
| | - Lingxue Tang
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical UniversityHefei, Anhui, China
- Department of Oncology, Anhui Medical UniversityHefei, Anhui, China
| | - Sheng Yu
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical UniversityHefei, Anhui, China
- Department of Oncology, Anhui Medical UniversityHefei, Anhui, China
| | - Anlong Li
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical UniversityHefei, Anhui, China
- Department of Oncology, Anhui Medical UniversityHefei, Anhui, China
| | - Han Ge
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical UniversityHefei, Anhui, China
- Department of Oncology, Anhui Medical UniversityHefei, Anhui, China
| | - Runze Huang
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical UniversityHefei, Anhui, China
- Department of Oncology, Anhui Medical UniversityHefei, Anhui, China
| | - Huaidong Cheng
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical UniversityHefei, Anhui, China
- Department of Oncology, Anhui Medical UniversityHefei, Anhui, China
- Department of Oncology, Shenzhen Hospital of Southern Medical UniversityShenzhen, Guangdong, China
| |
Collapse
|
6
|
Banks WA, Hansen KM, Erickson MA, Crews FT. High-mobility group box 1 (HMGB1) crosses the BBB bidirectionally. Brain Behav Immun 2023; 111:386-394. [PMID: 37146655 DOI: 10.1016/j.bbi.2023.04.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 04/25/2023] [Accepted: 04/30/2023] [Indexed: 05/07/2023] Open
Abstract
High-mobility group box 1 (HMGB1) is a ubiquitous protein that regulates transcription in the nucleus, and is an endogenous damage-associated molecular pattern molecule that activates the innate immune system. HMGB1 activates the TLR4 and RAGE recepto, inducing downstream signals reminiscent of cytokines that have been found to cross the blood-brain barrier (BBB). Blood HMGB1 increases in stroke, sepsis, senescence, alcohol binge drinking and other conditions. Here, we examined the ability of HMGB1 radioactively labeled with iodine (I-HMGB1) to cross the BBB. We found that I-HMGB1 readily entered into mouse brain from the circulation with a unidirectional influx rate of 0.654 μl/g-min. All brain regions tested took up I-HMGB1; uptake was greatest by the olfactory bulb and least in the striatum. Transport was not reliably inhibited by unlabeled HMGB1 nor by inhibitors of TLR4, TLR2, RAGE, or CXCR4. Uptake was enhanced by co-injection of wheatgerm agglutinin, suggestive of involvement of absorptive transcytosis as a mechanism of transport. Induction of inflammation/neuroinflammation with lipopolysaccharide is known to increase blood HMGB1; we report here that brain transport is also increased by LPS-induced inflammation. Finally, we found that I-HMGB1 was also transported in the brain-to-blood direction, with both unlabeled HMGB1 or lipopolysaccharide increasing the transport rate. These results show that HMGB1 can bidirectionally cross the BBB and that those transport rates are enhanced by inflammation. Such transport provides a mechanism by which HMGB1 levels would impact neuroimmune signaling in both the brain and periphery.
Collapse
Affiliation(s)
- William A Banks
- Geriatric Research Educational and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, US State; Division of Gerontology and Geriatric Medicine, University of Washington School of Medicine, Seattle, WA, US State.
| | - Kim M Hansen
- Geriatric Research Educational and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, US State; Division of Gerontology and Geriatric Medicine, University of Washington School of Medicine, Seattle, WA, US State
| | - Michelle A Erickson
- Geriatric Research Educational and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, US State; Division of Gerontology and Geriatric Medicine, University of Washington School of Medicine, Seattle, WA, US State
| | - Fulton T Crews
- Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, US State
| |
Collapse
|
7
|
Chen H, She Q, Liu Y, Chen J, Qin Y, Lu C. The peripheral Epac1/p-Cav-1 pathway underlies the disruption of the vascular endothelial barrier following skin/muscle incision and retraction-induced chronic postsurgical pain. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:1377. [PMID: 36660643 PMCID: PMC9843368 DOI: 10.21037/atm-22-6069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 12/20/2022] [Indexed: 12/29/2022]
Abstract
Background Vascular endothelial barrier disruption is pivotal in the development of acute and chronic pain. Here, we demonstrate a previously unidentified molecular mechanism in which activation of the peripheral Epac1/p-Cav-1 pathway accelerated the disruption of the vascular endothelial barrier, thereby promoting chronic postsurgical pain (CPSP). Methods We established a rat model of CPSP induced by skin/muscle incision and retraction (SMIR). Pain behaviors were assessed by the mechanical withdrawal threshold (MWT) at different times. Local muscle tissues around the incision were isolated to detect the vascular permeability and the expression of Epac1 and Cav-1. They were assessed by western blot and immunofluorescence staining. Results SMIR increased vascular endothelial permeability and the number of macrophages and endothelial cells in the muscle tissues around the incision. The peripheral upregulation of Epac1 was macrophage-derived, whereas that of p-Cav-1 was both macrophage and endothelial cell-derived in the SMIR model. Moreover, the Epac1 agonist 8-pCPT could induce mechanical sensitivity, increase the expression of p-Cav-1, and disrupt vascular endothelial barrier in normal rats. The Epac1 inhibitor CE3F4 attenuated established SMIR-induced mechanical hyperalgesia, the upregulation of p-Cav-1 and vascular endothelial barrier. Finally, we showed that intrathecal injection of Cav-1siRNA relieved SMIR-induced mechanical allodynia, but had no effects of the expression of Epac1. Conclusions Collectively, these results revealed a molecular mechanism for modulating CPSP through the peripheral Epac1/Cav-1 pathway. Importantly, targeting Epac1/Cav-1 signaling might be a potential treatment for CPSP.
Collapse
Affiliation(s)
- Hongsheng Chen
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, China
| | - Qing She
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, China
| | - Yanfang Liu
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, China
| | - Junjie Chen
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, China
| | - Yibin Qin
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, China
| | - Cui'e Lu
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
8
|
Popov LD. Deciphering the relationship between caveolae-mediated intracellular transport and signalling events. Cell Signal 2022; 97:110399. [PMID: 35820545 DOI: 10.1016/j.cellsig.2022.110399] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/02/2022] [Accepted: 07/05/2022] [Indexed: 11/30/2022]
Abstract
The caveolae-mediated transport across polarized epithelial cell barriers has been largely deciphered in the last decades and is considered the second essential intracellular transfer mechanism, after the clathrin-dependent endocytosis. The basic cell biology knowledge was supplemented recently, with the molecular mechanisms beyond caveolae generation implying the key contribution of the lipid-binding proteins (the structural protein Caveolin and the adapter protein Cavin), along with the bulb coat stabilizing molecules PACSIN-2 and Eps15 homology domain protein-2. The current attention is focused also on caveolae architecture (such as the bulb coat, the neck, the membrane funnel inside the bulb, and the associated receptors), and their specific tasks during the intracellular transport of various cargoes. Here, we resume the present understanding of the assembly, detachment, and internalization of caveolae from the plasma membrane lipid raft domains, and give an updated view on transcytosis and endocytosis, the two itineraries of cargoes transport via caveolae. The review adds novel data on the signalling molecules regulating caveolae intracellular routes and on the transport dysregulation in diseases. The therapeutic possibilities offered by exploitation of Caveolin-1 expression and caveolae trafficking, and the urgent issues to be uncovered conclude the review.
Collapse
Affiliation(s)
- Lucia-Doina Popov
- Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, 8, B.P. Hasdeu Street, 050568 Bucharest, Romania.
| |
Collapse
|
9
|
Jones JH, Minshall RD. Endothelial Transcytosis in Acute Lung Injury: Emerging Mechanisms and Therapeutic Approaches. Front Physiol 2022; 13:828093. [PMID: 35431977 PMCID: PMC9008570 DOI: 10.3389/fphys.2022.828093] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 02/28/2022] [Indexed: 01/08/2023] Open
Abstract
Acute Lung Injury (ALI) is characterized by widespread inflammation which in its severe form, Acute Respiratory Distress Syndrome (ARDS), leads to compromise in respiration causing hypoxemia and death in a substantial number of affected individuals. Loss of endothelial barrier integrity, pneumocyte necrosis, and circulating leukocyte recruitment into the injured lung are recognized mechanisms that contribute to the progression of ALI/ARDS. Additionally, damage to the pulmonary microvasculature by Gram-negative and positive bacteria or viruses (e.g., Escherichia coli, SARS-Cov-2) leads to increased protein and fluid permeability and interstitial edema, further impairing lung function. While most of the vascular leakage is attributed to loss of inter-endothelial junctional integrity, studies in animal models suggest that transendothelial transport of protein through caveolar vesicles, known as transcytosis, occurs in the early phase of ALI/ARDS. Here, we discuss the role of transcytosis in healthy and injured endothelium and highlight recent studies that have contributed to our understanding of the process during ALI/ARDS. We also cover potential approaches that utilize caveolar transport to deliver therapeutics to the lungs which may prevent further injury or improve recovery.
Collapse
Affiliation(s)
- Joshua H. Jones
- Department of Pharmacology, University of Illinois College of Medicine at Chicago, Chicago, IL, United States
| | - Richard D. Minshall
- Department of Pharmacology, University of Illinois College of Medicine at Chicago, Chicago, IL, United States,Department of Anesthesiology, University of Illinois College of Medicine at Chicago, Chicago, IL, United States,*Correspondence: Richard D. Minshall,
| |
Collapse
|
10
|
Sun S, Cai B, Li Y, Su W, Zhao X, Gong B, Li Z, Zhang X, Wu Y, Chen C, Tsang SH, Yang J, Li X. HMGB1 and Caveolin-1 related to RPE cell senescence in age-related macular degeneration. Aging (Albany NY) 2020; 11:4323-4337. [PMID: 31284269 PMCID: PMC6660032 DOI: 10.18632/aging.102039] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 06/20/2019] [Indexed: 01/09/2023]
Abstract
Accumulation of lipofuscin in the retinal pigment epithelium (RPE) is considered a major cause of RPE dysfunction and senescence in age-related macular degeneration (AMD), and N-retinylidene-N-retinylethanolamine (A2E) is the main fluorophore identified in lipofuscin from aged human eyes. Here, human-induced pluripotent stem cell (iPSC)-RPE was generated from healthy individuals to reveal proteomic changes associated with A2E-related RPE cell senescence. A novel RPE cell senescence-related protein, high-mobility group box 1 (HMGB1), was identified based on proteomic mass spectrometry measurements on iPSC-RPE with A2E treatment. Furthermore, HMGB1 upregulated Caveolin-1, which also was related RPE cell senescence. To investigate whether changes in HMGB1 and Caveolin-1 expression under A2E exposure contribute to RPE cell senescence, human ARPE-19 cells were stimulated with A2E; expression of HMGB1, Caveolin-1, tight junction proteins and senescent phenotypes were verified. HMGB1 inhibition alleviated A2E induced cell senescence. Migration of RPE cells was evaluated. Notably, A2E less than or equal to 10μM induced both HMGB1 and Caveolin-1 protein upregulation and HMGB1 translocation, while Caveolin-1 expression was downregulated when there was more than 10μM A2E. Our data indicate that A2E-induced upregulation of HMGB1、Caveolin-1 and HMGB1 release may relate to RPE cell senescence and play a role in the pathogenesis of AMD.
Collapse
Affiliation(s)
- Shuo Sun
- Tianjin Key Laboratory of Retinal Functions and Diseases, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, People's Republic of China
| | - Bincui Cai
- Tianjin Key Laboratory of Retinal Functions and Diseases, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, People's Republic of China
| | - Yao Li
- Edward S. Harkness Eye Institute, New York-Presbyterian Hospital, New York, NY 10032, USA.,Departments of Ophthalmology, Columbia University, New York, NY 10027, USA
| | - Wenqi Su
- Tianjin Key Laboratory of Retinal Functions and Diseases, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, People's Republic of China
| | - Xuzheng Zhao
- Tangshan Eye Hospital, Tangshan, People's Republic of China
| | - Boteng Gong
- Tianjin Key Laboratory of Retinal Functions and Diseases, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, People's Republic of China
| | - Zhiqing Li
- Tianjin Key Laboratory of Retinal Functions and Diseases, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, People's Republic of China
| | - Xiaomin Zhang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, People's Republic of China
| | - Yalin Wu
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Eye Institute of Xiamen University, College of Medicine, Xiamen University, Xiamen City, People's Republic of China
| | - Chao Chen
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Eye Institute of Xiamen University, College of Medicine, Xiamen University, Xiamen City, People's Republic of China
| | - Stephen H Tsang
- Edward S. Harkness Eye Institute, New York-Presbyterian Hospital, New York, NY 10032, USA.,Departments of Ophthalmology, Columbia University, New York, NY 10027, USA
| | - Jin Yang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, People's Republic of China
| | - Xiaorong Li
- Tianjin Key Laboratory of Retinal Functions and Diseases, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, People's Republic of China
| |
Collapse
|
11
|
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
|
12
|
Tjakra M, Wang Y, Vania V, Hou Z, Durkan C, Wang N, Wang G. Overview of Crosstalk Between Multiple Factor of Transcytosis in Blood Brain Barrier. Front Neurosci 2020; 13:1436. [PMID: 32038141 PMCID: PMC6990130 DOI: 10.3389/fnins.2019.01436] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Accepted: 12/19/2019] [Indexed: 12/16/2022] Open
Abstract
Blood brain barrier (BBB) conserves unique regulatory system to maintain barrier tightness while allowing adequate transport between neurovascular units. This mechanism possess a challenge for drug delivery, while abnormality may result in pathogenesis. Communication between vascular and neural system is mediated through paracellular and transcellular (transcytosis) pathway. Transcytosis itself showed dependency with various components, focusing on caveolae-mediated. Among several factors, intense communication between endothelial cells, pericytes, and astrocytes is the key for a normal development. Regulatory signaling pathway such as VEGF, Notch, S1P, PDGFβ, Ang/Tie, and TGF-β showed interaction with the transcytosis steps. Recent discoveries showed exploration of various factors which has been proven to interact with one of the process of transcytosis, either endocytosis, endosomal rearrangement, or exocytosis. As well as providing a hypothetical regulatory pathway between each factors, specifically miRNA, mechanical stress, various cytokines, physicochemical, basement membrane and junctions remodeling, and crosstalk between developmental regulatory pathways. Finally, various hypotheses and probable crosstalk between each factors will be expressed, to point out relevant research application (Drug therapy design and BBB-on-a-chip) and unexplored terrain.
Collapse
Affiliation(s)
- Marco Tjakra
- Key Laboratory for Biorheological Science and Technology, Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
| | - Yeqi Wang
- Key Laboratory for Biorheological Science and Technology, Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
| | - Vicki Vania
- Key Laboratory for Biorheological Science and Technology, Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
| | - Zhengjun Hou
- Key Laboratory for Biorheological Science and Technology, Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
| | - Colm Durkan
- The Nanoscience Centre, University of Cambridge, Cambridge, United Kingdom
| | - Nan Wang
- The Nanoscience Centre, University of Cambridge, Cambridge, United Kingdom
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology, Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
| |
Collapse
|
13
|
Wang M, Gauthier A, Daley L, Dial K, Wu J, Woo J, Lin M, Ashby C, Mantell LL. The Role of HMGB1, a Nuclear Damage-Associated Molecular Pattern Molecule, in the Pathogenesis of Lung Diseases. Antioxid Redox Signal 2019; 31:954-993. [PMID: 31184204 PMCID: PMC6765066 DOI: 10.1089/ars.2019.7818] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 06/07/2019] [Indexed: 12/11/2022]
Abstract
Significance: High-mobility group protein box 1 (HMGB1), a ubiquitous nuclear protein, regulates chromatin structure and modulates the expression of many genes involved in the pathogenesis of lung cancer and many other lung diseases, including those that regulate cell cycle control, cell death, and DNA replication and repair. Extracellular HMGB1, whether passively released or actively secreted, is a danger signal that elicits proinflammatory responses, impairs macrophage phagocytosis and efferocytosis, and alters vascular remodeling. This can result in excessive pulmonary inflammation and compromised host defense against lung infections, causing a deleterious feedback cycle. Recent Advances: HMGB1 has been identified as a biomarker and mediator of the pathogenesis of numerous lung disorders. In addition, post-translational modifications of HMGB1, including acetylation, phosphorylation, and oxidation, have been postulated to affect its localization and physiological and pathophysiological effects, such as the initiation and progression of lung diseases. Critical Issues: The molecular mechanisms underlying how HMGB1 drives the pathogenesis of different lung diseases and novel therapeutic approaches targeting HMGB1 remain to be elucidated. Future Directions: Additional research is needed to identify the roles and functions of modified HMGB1 produced by different post-translational modifications and their significance in the pathogenesis of lung diseases. Such studies will provide information for novel approaches targeting HMGB1 as a treatment for lung diseases.
Collapse
Affiliation(s)
- Mao Wang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York
| | - Alex Gauthier
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York
| | - LeeAnne Daley
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York
| | - Katelyn Dial
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York
| | - Jiaqi Wu
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York
| | - Joanna Woo
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York
| | - Mosi Lin
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York
| | - Charles Ashby
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York
| | - Lin L. Mantell
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York
- Center for Inflammation and Immunology, The Feinstein Institute for Medical Research, Northwell Health, Manhasset, New York
| |
Collapse
|
14
|
Gu Y, Cai R, Zhang C, Xue Y, Pan Y, Wang J, Zhang Z. miR-132-3p boosts caveolae-mediated transcellular transport in glioma endothelial cells by targeting PTEN/PI3K/PKB/Src/Cav-1 signaling pathway. FASEB J 2018; 33:441-454. [PMID: 30024792 DOI: 10.1096/fj.201800095rr] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Blood-brain tumor barrier (BTB) impedes the transportation of antitumor therapeutic drugs into brain tumors. Its mechanism is still unknown, but learning how to improve the BTB permeability is critical for drug intervention. Recently, microRNAs (miRNAs) have appeared as regulation factors of numerous biologic processes and therapeutic targets of diverse diseases. In this study, we have identified that miR-132-3p is an essential miRNA by increasing the transcellular transport through the BTB. We found that miR-132-3p expression was significantly up-regulated in glioma endothelial cells (GECs). Furthermore we showed that miR132-3p+ greatly induced the endocytosis of cholera toxin subunit B and FITC-bovine serum albumin and up-regulated the expression of p-PKB, p-Src and Tyr14 phosphorylation of caveolin-1 (p-Cav-1), while phosphatase and tensin homolog deleted on chromosome 10 (PTEN) expression was markedly down-regulated in GECs. Our results identify PTEN as a direct and functional downstream target of miR-132-3p, which is involved in the regulation of p-PKB, p-Src, and p-Cav-1. The inhibitors for PI3K and Src significantly reversed the increase of p-Cav-1 induced by miR-132-3p. Moreover, overexpression of PTEN greatly reduced the endocytosis of cholera toxin subunit B and the up-regulation of p-Cav-1 induced by agomiR132-3p, suggesting that miR132-3p+ increases the endothelial permeability by inhibition of PTEN expression. In addition, miR132-3p+ significantly increased the delivery of doxorubicin across the BTB in vitro and contributed to the accumulation of doxorubicin within the brain tumor tissue. Our results show that miR-132-3p contributes to the increased permeability of BTB by targeting PTEN/PI3K/PKB/Src/Cav-1, thereby revealing a novel drug target for the treatment of brain gliomas.-Gu, Y., Cai, R., Zhang, C., Xue, Y., Pan, Y., Wang, J., Zhang, Z. miR-132-3p boosts caveolae-mediated transcellular transport in glioma endothelial cells by targeting PTEN/PI3K/PKB/Src/Cav-1 signaling pathway.
Collapse
Affiliation(s)
- Yanting Gu
- Department of Physiology, Life Science and Biopharmaceutical Institution, Shenyang Pharmaceutical University, Shenyang, China; and
| | - Ruiping Cai
- Department of Physiology, Life Science and Biopharmaceutical Institution, Shenyang Pharmaceutical University, Shenyang, China; and
| | - Cai Zhang
- Department of Physiology, Life Science and Biopharmaceutical Institution, Shenyang Pharmaceutical University, Shenyang, China; and
| | - Yixue Xue
- Department of Neurobiology, College Basic of Medicine, China Medical University, Shenyang, China
| | - Yali Pan
- Department of Physiology, Life Science and Biopharmaceutical Institution, Shenyang Pharmaceutical University, Shenyang, China; and
| | - Jiahong Wang
- Department of Physiology, Life Science and Biopharmaceutical Institution, Shenyang Pharmaceutical University, Shenyang, China; and
| | - Zhou Zhang
- Department of Physiology, Life Science and Biopharmaceutical Institution, Shenyang Pharmaceutical University, Shenyang, China; and
| |
Collapse
|
15
|
Zhou H, Jin C, Cui L, Xing H, Liu J, Liao W, Liao H, Yu Y. HMGB1 contributes to the irradiation-induced endothelial barrier injury through receptor for advanced glycation endproducts (RAGE). J Cell Physiol 2018; 233:6714-6721. [PMID: 29215715 DOI: 10.1002/jcp.26341] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 12/04/2017] [Indexed: 12/20/2022]
Abstract
This study aimed to investigate whether HMGB1 (high mobility group box-1 protein) and receptor for advanced glycation end products (RAGE) were involved in the irradiation-induced endothelial barrier damage and their mechanism. We constructed the damage model of endothelium barrier model with bEnd.3 cells. The permeability of endothelial barrier was detected by sodium fluorescein (Na-F) permeation test, and the irradiation dose which could induce permeability transition was determined by being exposed to different irradiation doses (5, 10, 15, 20 Gy). MTT assay was applied to detect cell viability under different concentrations of HMGB1, glycyrrhizic acid (GA, a specific inhibitor of HMGB1), and FPS-ZM1 (a blood-brain-barrier permeant blocker of RAGE V domain-mediated ligand binding). The expression of HMGB1, RAGE, and related molecules involved in MAPK signaling pathway, MMP-2, MMP-9, ZO-1, and claudin 5 of differently treated groups were measured by qRT-PCR, western blot, and immunofluorescence. Cells possessed stable endothelial barrier function on 4-7 days after seeded on transwell plates. The permeability of endothelial barrier would change under at least 10 Gy radiation. Both radiation and HMGB1 treatment alone could improve the permeability. After irradiation, the expressions of HMGB1 and RAGE increased and MAPK signal pathway was activated. Meanwhile, MMP-2 and MMP-9 were overexpressed, while the expression of tight junction proteins ZO-1 and claudin 5 was decreased. Radiation could activate MAPK signaling pathway through promoting the expression of HMGB1 and RAGE, which further led to endothelial barrier injury and changed its permeability.
Collapse
Affiliation(s)
- Haihong Zhou
- Department of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Congli Jin
- Department of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Lili Cui
- Department of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Huaijie Xing
- Department of Neurology, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Jun Liu
- Department of Neurology, SUN YAT-SEN Medical Hospital, SUN YAT-SEN University, Guangzhou, Guangdong, China
| | - Wang Liao
- Department of Neurology, SUN YAT-SEN Medical Hospital, SUN YAT-SEN University, Guangzhou, Guangdong, China
| | - Haojie Liao
- Department of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Yangsheng Yu
- Department of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| |
Collapse
|
16
|
Xu Q, Bo L, Hu J, Geng J, Chen Y, Li X, Chen F, Song J. High mobility group box 1 was associated with thrombosis in patients with atrial fibrillation. Medicine (Baltimore) 2018; 97:e0132. [PMID: 29595637 PMCID: PMC5895421 DOI: 10.1097/md.0000000000010132] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION High mobility group box 1 (HMGB1) is a member of the HMGB family that is involved in inflammatory disease-related thrombosis. We hypothesize that HMGB1 and its downstream factors are associated with thrombosis in atrial fibrillation (AF). MATERIALS AND METHODS Our experimental materials were the left atrial appendage (LAA) tissues from patients undergoing valve replacement. The samples were divided into 3 groups: a sinus rhythm group (n = 15), an AF(+)thrombus(-)group (n = 15), and an AF(+) thrombus (+)group (n = 15). The expression of HMGB1, Toll-like receptor 4 (TLR4), advanced glycation end product (RAGE), myeloid differentiation factor 88 (MyD88), nuclear factor κB (NFκB), p-NFκB, and tissue factor (TF) were detected by Western blot and immunohistochemical (IHC) staining. The expressions of interleukin-1 beta, interleukin 6, and tumor necrosis factor-alpha were detected by quantitative real-time PCR. RESULTS The Western blots revealed significantly higher expressions of HMGB1, MyD88, p-NFκB/NFκB, and TF in the AF(+)thrombus(+) group than in the other 2 groups. However, no differences in TLR4 or RAGE expression were found between the groups. IHC staining also revealed higher expressions of HMGB1 and TF in the AF(+)thrombus(+) group. The increased mRNA expressions of classic inflammatory factors (i.e., interleukin-1 beta, interleukin 6, and tumor necrosis factor-alpha) in AF(+)thrombus(+) group further validated the correlation between inflammation and thrombi in atrial fibrillation. CONCLUSIONS HMGB1 was associated with thrombosis in patients with AF via the MyD88/NFκB pathway after adjustment for cardiac and extra cardiac inflammation variables.
Collapse
Affiliation(s)
- Qiwen Xu
- Department of Cardiology, Nanjing Drum Tower Hospital, Nanjing Medical University
| | - Lin Bo
- Department of Cardiology, Nanjing Drum Tower Hospital, Nanjing Medical University
| | - Jiaxin Hu
- Department of Cardiology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Jin Geng
- Department of Cardiology, Nanjing Drum Tower Hospital, Nanjing Medical University
| | - Yuhan Chen
- Department of Cardiology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Xuelin Li
- Department of Cardiology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Fu Chen
- Department of Cardiology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Jie Song
- Department of Cardiology, Nanjing Drum Tower Hospital, Nanjing Medical University
| |
Collapse
|
17
|
Transcytosis Involvement in Transport System and Endothelial Permeability of Vascular Leakage during Dengue Virus Infection. Viruses 2018; 10:v10020069. [PMID: 29419739 PMCID: PMC5850376 DOI: 10.3390/v10020069] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 01/19/2018] [Accepted: 02/01/2018] [Indexed: 12/19/2022] Open
Abstract
The major role of endothelial cells is to maintain homeostasis of vascular permeability and to preserve the integrity of vascular vessels to prevent fluid leakage. Properly functioning endothelial cells promote physiological balance and stability for blood circulation and fluid components. A monolayer of endothelial cells has the ability to regulate paracellular and transcellular pathways for transport proteins, solutes, and fluid. In addition to the paracellular pathway, the transcellular pathway is another route of endothelial permeability that mediates vascular permeability under physiologic conditions. The transcellular pathway was found to be associated with an assortment of disease pathogeneses. The clinical manifestation of severe dengue infection in humans is vascular leakage and hemorrhagic diatheses. This review explores and describes the transcellular pathway, which is an alternate route of vascular permeability during dengue infection that corresponds with the pathologic finding of intact tight junction. This pathway may be the route of albumin transport that causes endothelial dysfunction during dengue virus infection.
Collapse
|
18
|
Parthasarathi K. The Pulmonary Vascular Barrier: Insights into Structure, Function, and Regulatory Mechanisms. MOLECULAR AND FUNCTIONAL INSIGHTS INTO THE PULMONARY VASCULATURE 2018; 228:41-61. [DOI: 10.1007/978-3-319-68483-3_3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
19
|
Corrêa LH, Corrêa R, Farinasso CM, de Sant'Ana Dourado LP, Magalhães KG. Adipocytes and Macrophages Interplay in the Orchestration of Tumor Microenvironment: New Implications in Cancer Progression. Front Immunol 2017; 8:1129. [PMID: 28970834 PMCID: PMC5609576 DOI: 10.3389/fimmu.2017.01129] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 08/28/2017] [Indexed: 12/22/2022] Open
Abstract
Inflammation has been known as one of the main keys to the establishment and progression of cancers. Chronic low-grade inflammation is also a strategic condition that underlies the causes and development of metabolic syndrome and obesity. Moreover, obesity has been largely related to poor prognosis of tumors by modulating tumor microenvironment with secretion of several inflammatory mediators by tumor-associated adipocytes (TAAs), which can modulate and recruit tumor-associated macrophages. Thus, the understanding of cellular and molecular mechanisms that underlay and link inflammation, obesity, and cancer is crucial to identify potential targets that interfere with this important route. Knowledge about the exact role of each component of the tumor microenvironment is not yet fully understood, but the new insights in literature highlight the essential role of adipocytes and macrophages interplay as key factor to determine the fate of cancer progression. In this review article, we focus on the functions of adipocytes and macrophages orchestrating cellular and molecular mechanisms that lead to inflammatory modulation in tumor microenvironment, which will be crucial to cancer establishment. We also emphasized the mechanisms by which the tumor promotes itself by recruiting and polarizing macrophages, discussing the role of adipocytes in this process. In addition, we discuss here the newest possible anticancer therapeutic treatments aiming to retard the development of the tumor based on what is known about cancer, adipocyte, and macrophage polarization.
Collapse
Affiliation(s)
- Luís Henrique Corrêa
- Laboratory of Immunology and Inflammation, Department of Cell Biology, University of Brasilia, Brasilia, Brazil
| | - Rafael Corrêa
- Laboratory of Immunology and Inflammation, Department of Cell Biology, University of Brasilia, Brasilia, Brazil
| | - Cecília Menezes Farinasso
- Laboratory of Immunology and Inflammation, Department of Cell Biology, University of Brasilia, Brasilia, Brazil
| | | | - Kelly Grace Magalhães
- Laboratory of Immunology and Inflammation, Department of Cell Biology, University of Brasilia, Brasilia, Brazil
| |
Collapse
|
20
|
Intrinsically Disordered Regions in Serum Albumin: What Are They For? Cell Biochem Biophys 2017; 76:39-57. [PMID: 28281231 DOI: 10.1007/s12013-017-0785-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 02/13/2017] [Indexed: 12/16/2022]
Abstract
Serum albumin is a major plasma protein in mammalian blood. The importance of this protein lies in its roles in both bioregulation and transport phenomena. Serum albumin binds various metal ions and participates in the transport and storage of fatty acids, bilirubin, steroids amino acids, and many other ligands, usually with regions of hydrophobic surface. Although the primary role of serum albumin is to transport various ligand, its versatile binding capacities and high concentration mean that it can assume a number of additional functions. The major goal of this article is to show how intrinsic disorder is encoded in the amino acid sequence of serum albumin, and how intrinsic disorder is related to functions of this important serum protein.
Collapse
|
21
|
Coccolini F, Acocella F, Morosi L, Brizzola S, Ghiringhelli M, Ceresoli M, Davoli E, Ansaloni L, D'Incalci M, Zucchetti M. High Penetration of Paclitaxel in Abdominal Wall of Rabbits after Hyperthermic Intraperitoneal Administration of Nab-Paclitaxel Compared to Standard Paclitaxel Formulation. Pharm Res 2017; 34:1180-1186. [PMID: 28247168 DOI: 10.1007/s11095-017-2132-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 02/20/2017] [Indexed: 01/30/2023]
Abstract
PURPOSE Paclitaxel (PTX) is currently used in combination with cisplatin for Hyperthermic Intraperitoneal Chemotherapy (HIPEC) for the treatment of peritoneal carcinomatosis. Albumin-bound PTX is a promising new drug for HIPEC because of its easy solubility in aqueous perfusion medium and possibly because of the tendency of albumin to cross physiological barriers and accumulate in tumor tissue. METHODS We tested the feasibility of using nab-paclitaxel in rabbits treated by HIPEC for 60 min compared with the classical formulation at an equivalent PTX dose. Samples of perfusate and blood were collected at different time points and peritoneal tissues were collected at the end of perfusion. PTX concentrations were determined by HPLC. The depth of paclitaxel penetration through the peritoneal barrier was assessed by mass spectrometry imaging. RESULTS PTX after nab-paclitaxel treatment penetrated up to 0.63 mm in the peritoneal wall, but after CRE-paclitaxel, it was not detectable in the peritoneum. Moreover, the peritoneal concentration after nab-paclitaxel was five times that after paclitaxel classical formulation. Despite the high levels reached in the peritoneum, systemic exposure of PTX was low. CONCLUSIONS Our results show that nab-paclitaxel penetrates into the abdominal wall better than CRE-paclitaxel, in terms of effective penetration and peritoneal tissue concentration.
Collapse
Affiliation(s)
- Federico Coccolini
- General, Emergency and Trauma Surgery Department Papa Giovanni XXIII hospital, Bergamo, Italy
| | - Fabio Acocella
- Dipartimento di Scienze Veterinarie per la Salute, la Produzione Animale e la Sicurezza Alimentare, Università degli Studi di Milano, Milan, Italy
| | - Lavinia Morosi
- Oncology Department, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy.
| | - Stefano Brizzola
- Dipartimento di Scienze Veterinarie per la Salute, la Produzione Animale e la Sicurezza Alimentare, Università degli Studi di Milano, Milan, Italy
| | - Matteo Ghiringhelli
- Dipartimento di Scienze Veterinarie per la Salute, la Produzione Animale e la Sicurezza Alimentare, Università degli Studi di Milano, Milan, Italy
| | - Marco Ceresoli
- General, Emergency and Trauma Surgery Department Papa Giovanni XXIII hospital, Bergamo, Italy
| | - Enrico Davoli
- Mass Spectrometry Laboratory, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Luca Ansaloni
- General, Emergency and Trauma Surgery Department Papa Giovanni XXIII hospital, Bergamo, Italy
| | - Maurizio D'Incalci
- Oncology Department, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Massimo Zucchetti
- Oncology Department, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| |
Collapse
|
22
|
Gillrie MR, Ho M. Dynamic interactions of Plasmodium spp. with vascular endothelium. Tissue Barriers 2017; 5:e1268667. [PMID: 28452684 PMCID: PMC5362994 DOI: 10.1080/21688370.2016.1268667] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 11/24/2016] [Accepted: 11/30/2016] [Indexed: 12/18/2022] Open
Abstract
Plasmodial species are protozoan parasites that infect erythrocytes. As such, they are in close contact with microvascular endothelium for most of the life cycle in the mammalian host. The host-parasite interactions of this stage of the infection are responsible for the clinical manifestations of the disease that range from a mild febrile illness to severe and frequently fatal syndromes such as cerebral malaria and multi-organ failure. Plasmodium falciparum, the causative agent of the most severe form of malaria, is particularly predisposed to modulating endothelial function through either direct adhesion to endothelial receptor molecules, or by releasing potent host and parasite products that can stimulate endothelial activation and/or disrupt barrier function. In this review, we provide a critical analysis of the current clinical and laboratory evidence for endothelial dysfunction during severe P. falciparum malaria. Future investigations using state-of-the-art technologies such as mass cytometry and organs-on-chips to further delineate parasite-endothelial cell interactions are also discussed.
Collapse
Affiliation(s)
- Mark R. Gillrie
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, Alberta, Canada
- Department of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - May Ho
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|