1
|
Xie R, Yang Y, Jiang X, Gao L, Sun J, Yang J. The effect of modulating platelet reactive oxygen species by the addition of antioxidants to prevent clearance of cold-stored platelets. Hematol Transfus Cell Ther 2024; 46 Suppl 6:S272-S283. [PMID: 39500660 PMCID: PMC11726102 DOI: 10.1016/j.htct.2024.09.2479] [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: 05/13/2024] [Accepted: 09/06/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND It is known that the rapid clearance of cold-stored platelets is attributed to various storage lesions, including an abnormal increase in reactive oxygen species when platelets are exposed to cold temperatures. As an antioxidant, N-acetylcysteine exhibits some significant effects on scavenging various reactive oxygen species and inhibiting cell damage and apoptosis. AIMS This study aimed to investigate the effects of N-acetylcysteine on reducing reactive oxygen species production and protecting cold-stored platelets from phagocytosis and clearance, and to determine the optimal concentration of N-acetylcysteine. METHODS Platelet concentrates were divided into three groups: room-temperature-stored platelets, cold-stored platelets, and cold-stored platelets with the addition of different concentrations of N-acetylcysteine. After five days of storage, reactive oxygen species production, lipid peroxidation levels, activation marker expressions, GPIb/ɑ desialylation with exposure of glycan residues and other quality parameters of platelets were measured and compared between the groups. Phagocytosis of platelets was detected by phorbol 12-myristate 13-acetate-activated THP-1 or Hep G2 cells. Moreover, the recovery of infused platelets was measured in severe combined immunodeficient mice at different timepoints. RESULTS After 5 days of storage, cytoplasmic reactive oxygen species significantly increased in chilled compared to non-chilled platelets; they were notably reduced with the addition of N-acetylcysteine, particularly at a concentration of 5 mM. Compared with chilled platelets, the P-selectin and phosphatidylserine expressions, as well as exposure of GPIb/ɑ glycan residues, were significantly reduced with 5 mM of N-acetylcysteine. Phagocytosis of platelets by THP-1 or Hep G2 cells was significantly lower in 5 mM of N-acetylcysteine compared to cold-stored platelets without N-acetylcysteine. CONCLUSIONS This study demonstrated correlations between reactive oxygen species production and their pro-oxidant effects on platelet clearance after cold storage. The addition of N-acetylcysteine at an appropriate concentration do not only protects chilled platelets from storage lesions caused by reactive oxygen species overproduction but also prevents platelet phagocytosis in vitro and clearance in vivo, thereby extending circulating time.
Collapse
Affiliation(s)
- Rufeng Xie
- Blood Engineering Laboratory, Shanghai Blood Center, Shanghai, China
| | - Yiming Yang
- Blood Engineering Laboratory, Shanghai Blood Center, Shanghai, China
| | - Xueyu Jiang
- Blood Engineering Laboratory, Shanghai Blood Center, Shanghai, China
| | - Li Gao
- Blood Engineering Laboratory, Shanghai Blood Center, Shanghai, China
| | - Juan Sun
- Blood Engineering Laboratory, Shanghai Blood Center, Shanghai, China
| | - Jie Yang
- Blood Engineering Laboratory, Shanghai Blood Center, Shanghai, China.
| |
Collapse
|
2
|
de Castro Sampaio SS, Ramalho MCC, de Souza CS, de Almeida Rodrigues B, de Mendonça GRS, Lazarini M. RHO subfamily of small GTPases in the development and function of hematopoietic cells. J Cell Physiol 2024:e31469. [PMID: 39434451 DOI: 10.1002/jcp.31469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/16/2024] [Accepted: 10/03/2024] [Indexed: 10/23/2024]
Abstract
RHOA, RHOB, and RHOC comprise a subfamily of RHO GTPase proteins famed for controlling cytoskeletal dynamics. RHO proteins operate downstream of multiple signals emerging from the microenvironment, leading to diverse cell responses, such as proliferation, adhesion, and migration. Therefore, RHO signaling has been centrally placed in the regulation of blood cells. Despite their high homology, unique roles of RHOA, RHOB, and RHOC have been described in hematopoietic cells. In this article, we overview the contribution of RHO proteins in the development and function of each blood cell lineage. Additionally, we highlight the aberrations of the RHO signaling pathways found in hematological malignancies, providing clues for the identification of new therapeutic targets.
Collapse
Affiliation(s)
| | | | - Caroline Santos de Souza
- Department of Clinical and Experimental Oncology, Federal University of São Paulo, São Paulo, Brazil
| | | | | | - Mariana Lazarini
- Department of Clinical and Experimental Oncology, Federal University of São Paulo, São Paulo, Brazil
| |
Collapse
|
3
|
Wei C, Zhang J, Peng S, Liu J, Xu Y, Zhao M, Xu S, Pan W, Yin Z, Zheng Z, Qin JJ, Wan J, Wang M. Resolvin D1 attenuates Ang II-induced hypertension in mice by inhibiting the proliferation, migration and phenotypic transformation of vascular smooth muscle cells by blocking the RhoA/mitogen-activated protein kinase pathway. J Hypertens 2024; 42:420-431. [PMID: 37937508 PMCID: PMC10842678 DOI: 10.1097/hjh.0000000000003610] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 10/16/2023] [Accepted: 10/17/2023] [Indexed: 11/09/2023]
Abstract
The proliferation, migration and phenotypic transformation of vascular smooth muscle cells contribute to vascular remodeling and hypertension. Resolvin D1 (RvD1) is a specialized pro-resolving lipid mediator that has been shown to have anti-inflammatory effects and can protect against different cardiovascular diseases. However, the role and mechanism of RvD1 in hypertension are not clear. The current study investigated the role of RvD1 in Ang II-induced hypertensive mice and Ang II-stimulated rat vascular smooth muscle cells. The results showed that RvD1 treatment significantly attenuated hypertension and vascular remodeling, as indicated by decreases in blood pressure, aortic media thickness and collagen deposition. In addition, RvD1 inhibited the proliferation, migration and phenotypic transformation of vascular smooth muscle cells (VSMCs) in vivo and in vitro . Notably, the protective effects of RvD1 were mediated by the Ras homolog gene family member A (RhoA)/mitogen-activated protein kinase (MAPK) signaling pathway. In conclusion, our findings demonstrated the potential benefits of RvD1 as a promising therapeutic agent in the treatment of vascular remodeling and hypertension.
Collapse
Affiliation(s)
- Cheng Wei
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University
- Cardiovascular Research Institute, Wuhan University
- Hubei Key Laboratory of Cardiology
| | - Jishou Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University
- Cardiovascular Research Institute, Wuhan University
- Hubei Key Laboratory of Cardiology
| | - Shanshan Peng
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University
- Cardiovascular Research Institute, Wuhan University
- Hubei Key Laboratory of Cardiology
| | - Jianfang Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University
- Cardiovascular Research Institute, Wuhan University
- Hubei Key Laboratory of Cardiology
| | - Yao Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University
- Cardiovascular Research Institute, Wuhan University
- Hubei Key Laboratory of Cardiology
| | - Mengmeng Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University
- Cardiovascular Research Institute, Wuhan University
- Hubei Key Laboratory of Cardiology
| | - Shuwan Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University
- Cardiovascular Research Institute, Wuhan University
- Hubei Key Laboratory of Cardiology
| | - Wei Pan
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University
- Cardiovascular Research Institute, Wuhan University
- Hubei Key Laboratory of Cardiology
| | - Zheng Yin
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University
- Cardiovascular Research Institute, Wuhan University
- Hubei Key Laboratory of Cardiology
| | - Zihui Zheng
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University
- Cardiovascular Research Institute, Wuhan University
- Hubei Key Laboratory of Cardiology
| | - Juan-Juan Qin
- Department of Geriatrics, Zhongnan Hospital of Wuhan University
- Center for Healthy Aging, Wuhan University School of Nursing, Wuhan, PR China
| | - Jun Wan
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University
- Cardiovascular Research Institute, Wuhan University
- Hubei Key Laboratory of Cardiology
| | - Menglong Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University
- Cardiovascular Research Institute, Wuhan University
- Hubei Key Laboratory of Cardiology
| |
Collapse
|
4
|
Pan D, Ladds G, Rahman KM, Pitchford SC. Exploring bias in platelet P2Y 1 signalling: Host defence versus haemostasis. Br J Pharmacol 2024; 181:580-592. [PMID: 37442808 PMCID: PMC10952580 DOI: 10.1111/bph.16191] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 04/21/2023] [Accepted: 06/22/2023] [Indexed: 07/15/2023] Open
Abstract
Platelets are necessary for maintaining haemostasis. Separately, platelets are important for the propagation of inflammation during the host immune response against infection. The activation of platelets also causes inappropriate inflammation in various disease pathologies, often in the absence of changes to haemostasis. The separate functions of platelets during inflammation compared with haemostasis are therefore varied and this will be reflected in distinct pathways of activation. The activation of platelets by the nucleotide adenosine diphosphate (ADP) acting on P2Y1 and P2Y12 receptors is important for the development of platelet thrombi during haemostasis. However, P2Y1 stimulation of platelets is also important during the inflammatory response and paradoxically in scenarios where no changes to haemostasis and platelet aggregation occur. In these events, Rho-GTPase signalling, rather than the canonical phospholipase Cβ (PLCβ) signalling pathway, is necessary. We describe our current understanding of these differences, reflecting on recent advances in knowledge of P2Y1 structure, and the possibility of biased agonism occurring from activation via other endogenous nucleotides compared with ADP. Knowledge arising from these different pathways of P2Y1 stimulation of platelets during inflammation compared with haemostasis may help therapeutic control of platelet function during inflammation or infection, while preserving essential haemostasis. LINKED ARTICLES: This article is part of a themed issue on Platelet purinergic receptor and non-thrombotic disease. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v181.4/issuetoc.
Collapse
Affiliation(s)
- Dingxin Pan
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical ScienceKing's College LondonLondonUK
| | - Graham Ladds
- Department of PharmacologyUniversity of CambridgeCambridgeUK
| | - Khondaker Miraz Rahman
- Chemical Biology Group, Institute of Pharmaceutical ScienceKing's College LondonLondonUK
| | - Simon C. Pitchford
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical ScienceKing's College LondonLondonUK
| |
Collapse
|
5
|
Arkless KL, Pan D, Shankar‐Hari M, Amison RT, Page CP, Rahman KM, Pitchford SC. Stimulation of platelet P2Y 1 receptors by different endogenous nucleotides leads to functional selectivity via biased signalling. Br J Pharmacol 2024; 181:564-579. [PMID: 36694432 PMCID: PMC10952403 DOI: 10.1111/bph.16039] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 12/13/2022] [Accepted: 01/13/2023] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND AND PURPOSE Platelet function during inflammation is dependent on activation by endogenous nucleotides. Non-canonical signalling via the P2Y1 receptor is important for these non-thrombotic functions of platelets. However, apart from ADP, the role of other endogenous nucleotides acting as agonists at P2Y1 receptors is unknown. This study compared the effects of ADP, Ap3A, NAD+ , ADP-ribose, and Up4A on platelet functions contributing to inflammation or haemostasis. EXPERIMENTAL APPROACH Platelets obtained from healthy human volunteers were incubated with ADP, Ap3A, NAD+ , ADP-ribose, or Up4A, with aggregation and fibrinogen binding measured (examples of function during haemostasis) or before exposure to fMLP to measure platelet chemotaxis (an inflammatory function). In silico molecular docking of these nucleotides to the binding pocket of P2Y1 receptors was then assessed. KEY RESULTS Platelet aggregation and binding to fibrinogen induced by ADP was not mimicked by NAD+ , ADP-ribose, and Up4A. However, these endogenous nucleotides induced P2Y1 -dependent platelet chemotaxis, an effect that required RhoA and Rac-1 activity, but not canonical PLC activity. Analysis of molecular docking of the P2Y1 receptor revealed distinct differences of amino acid interactions and depth of fit within the binding pocket for Ap3A, NAD+ , ADP-ribose, or Up4A compared with ADP. CONCLUSION AND IMPLICATIONS Platelet function (aggregation vs motility) can be differentially modulated by biased-agonist activation of P2Y1 receptors. This may be due to the character of the ligand-binding pocket interaction. This has implications for future therapeutic strategies aimed to suppress platelet activation during inflammation without affecting haemostasis as is the requirement of current ant-platelet drugs. LINKED ARTICLES This article is part of a themed issue on Platelet purinergic receptor and non-thrombotic disease. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v181.4/issuetoc.
Collapse
Affiliation(s)
- Kate L. Arkless
- Sackler Institute of Pulmonary PharmacologyInstitute of Pharmaceutical Science, King's College LondonLondonUK
| | - Dingxin Pan
- Sackler Institute of Pulmonary PharmacologyInstitute of Pharmaceutical Science, King's College LondonLondonUK
| | - Manu Shankar‐Hari
- School of Immunology and Microbial SciencesKing's College LondonLondonUK
- Centre for Inflammation ResearchThe University of EdinburghEdinburghUK
| | - Richard T. Amison
- Sackler Institute of Pulmonary PharmacologyInstitute of Pharmaceutical Science, King's College LondonLondonUK
| | - Clive P. Page
- Sackler Institute of Pulmonary PharmacologyInstitute of Pharmaceutical Science, King's College LondonLondonUK
| | - Khondaker Miraz Rahman
- Chemical Biology Group, Institute of Pharmaceutical ScienceKing's College LondonLondonUK
| | - Simon C. Pitchford
- Sackler Institute of Pulmonary PharmacologyInstitute of Pharmaceutical Science, King's College LondonLondonUK
| |
Collapse
|
6
|
Zhang Y, Zhao X, Shen B, Bai Y, Chang C, Stojanovic A, Wang C, Mack A, Deng G, Skidgel RA, Cheng N, Du X. Integrin β 3 directly inhibits the Gα 13-p115RhoGEF interaction to regulate G protein signaling and platelet exocytosis. Nat Commun 2023; 14:4966. [PMID: 37587112 PMCID: PMC10432399 DOI: 10.1038/s41467-023-40531-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 07/28/2023] [Indexed: 08/18/2023] Open
Abstract
The integrins and G protein-coupled receptors are both fundamental in cell biology. The cross talk between these two, however, is unclear. Here we show that β3 integrins negatively regulate G protein-coupled signaling by directly inhibiting the Gα13-p115RhoGEF interaction. Furthermore, whereas β3 deficiency or integrin antagonists inhibit integrin-dependent platelet aggregation and exocytosis (granule secretion), they enhance G protein-coupled RhoA activation and integrin-independent secretion. In contrast, a β3-derived Gα13-binding peptide or Gα13 knockout inhibits G protein-coupled RhoA activation and both integrin-independent and dependent platelet secretion without affecting primary platelet aggregation. In a mouse model of myocardial ischemia/reperfusion injury in vivo, the β3-derived Gα13-binding peptide inhibits platelet secretion of granule constituents, which exacerbates inflammation and ischemia/reperfusion injury. These data establish crucial integrin-G protein crosstalk, providing a rationale for therapeutic approaches that inhibit exocytosis in platelets and possibly other cells without adverse effects associated with loss of cell adhesion.
Collapse
Affiliation(s)
- Yaping Zhang
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Xiaojuan Zhao
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Bo Shen
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Yanyan Bai
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Claire Chang
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Aleksandra Stojanovic
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
- Dupage Medical Technology, Inc., Chicago, IL, 60612, USA
| | - Can Wang
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Andrew Mack
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Gary Deng
- Eli Lilly, Indianapolis, IN, 46285, USA
| | | | - Ni Cheng
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Xiaoping Du
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA.
| |
Collapse
|
7
|
Ribeiro V, Martins SG, Lopes AS, Thorsteinsdóttir S, Zilhão R, Carlos AR. NFIXing Cancer: The Role of NFIX in Oxidative Stress Response and Cell Fate. Int J Mol Sci 2023; 24:ijms24054293. [PMID: 36901722 PMCID: PMC10001739 DOI: 10.3390/ijms24054293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 02/15/2023] [Accepted: 02/20/2023] [Indexed: 02/24/2023] Open
Abstract
NFIX, a member of the nuclear factor I (NFI) family of transcription factors, is known to be involved in muscle and central nervous system embryonic development. However, its expression in adults is limited. Similar to other developmental transcription factors, NFIX has been found to be altered in tumors, often promoting pro-tumorigenic functions, such as leading to proliferation, differentiation, and migration. However, some studies suggest that NFIX can also have a tumor suppressor role, indicating a complex and cancer-type dependent role of NFIX. This complexity may be linked to the multiple processes at play in regulating NFIX, which include transcriptional, post-transcriptional, and post-translational processes. Moreover, other features of NFIX, including its ability to interact with different NFI members to form homodimers or heterodimers, therefore allowing the transcription of different target genes, and its ability to sense oxidative stress, can also modulate its function. In this review, we examine different aspects of NFIX regulation, first in development and then in cancer, highlighting the important role of NFIX in oxidative stress and cell fate regulation in tumors. Moreover, we propose different mechanisms through which oxidative stress regulates NFIX transcription and function, underlining NFIX as a key factor for tumorigenesis.
Collapse
Affiliation(s)
- Vanessa Ribeiro
- cE3c-CHANGE, Department of Animal Biology, Faculty of Sciences, University of Lisbon, 1749-016 Lisbon, Portugal
| | - Susana G. Martins
- cE3c-CHANGE, Department of Animal Biology, Faculty of Sciences, University of Lisbon, 1749-016 Lisbon, Portugal
| | - Ana Sofia Lopes
- cE3c-CHANGE, Department of Animal Biology, Faculty of Sciences, University of Lisbon, 1749-016 Lisbon, Portugal
- Centro Hospitalar de Lisboa Ocidental (CHLO), 1449-005 Lisbon, Portugal
| | - Sólveig Thorsteinsdóttir
- cE3c-CHANGE, Department of Animal Biology, Faculty of Sciences, University of Lisbon, 1749-016 Lisbon, Portugal
| | - Rita Zilhão
- cE3c-CHANGE, Department of Plant Biology, Faculty of Sciences, University of Lisbon, 1749-016 Lisbon, Portugal
| | - Ana Rita Carlos
- cE3c-CHANGE, Department of Animal Biology, Faculty of Sciences, University of Lisbon, 1749-016 Lisbon, Portugal
- Correspondence:
| |
Collapse
|
8
|
Structure-Activity Relationship Analysis of Rhosin, a RhoA GTPase Inhibitor, Reveals a New Class of Antiplatelet Agents. Int J Mol Sci 2023; 24:ijms24044167. [PMID: 36835579 PMCID: PMC9961652 DOI: 10.3390/ijms24044167] [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: 01/23/2023] [Revised: 02/14/2023] [Accepted: 02/17/2023] [Indexed: 02/22/2023] Open
Abstract
Current antiplatelet therapies have several clinical complications and are mostly irreversible in terms of suppressing platelet activity; hence, there is a need to develop improved therapeutic agents. Previous studies have implicated RhoA in platelet activation. Here, we further characterized the lead RhoA inhibitor, Rhosin/G04, in platelet function and present structure-activity relationship (SAR) analysis. A screening for Rhosin/G04 analogs in our chemical library by similarity and substructure searches revealed compounds that showed enhanced antiplatelet activity and suppressed RhoA activity and signaling. A screening for Rhosin/G04 analogs in our chemical library using similarity and substructure searches revealed compounds that showed enhanced antiplatelet activity and suppressed RhoA activity and signaling. SAR analysis revealed that the active compounds have a quinoline group optimally attached to the hydrazine at the 4-position and halogen substituents at the 7- or 8-position. Having indole, methylphenyl, or dichloro-phenyl substituents led to better potency. Rhosin/G04 contains a pair of enantiomers, and S-G04 is significantly more potent than R-G04 in inhibiting RhoA activation and platelet aggregation. Furthermore, the inhibitory effect is reversible, and S-G04 is capable of inhibiting diverse-agonist-stimulated platelet activation. This study identified a new generation of small-molecule RhoA inhibitors, including an enantiomer capable of broadly and reversibly modulating platelet activity.
Collapse
|
9
|
Dandamudi A, Akbar H, Cancelas J, Zheng Y. Rho GTPase Signaling in Platelet Regulation and Implication for Antiplatelet Therapies. Int J Mol Sci 2023; 24:ijms24032519. [PMID: 36768837 PMCID: PMC9917354 DOI: 10.3390/ijms24032519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 01/16/2023] [Accepted: 01/21/2023] [Indexed: 01/31/2023] Open
Abstract
Platelets play a vital role in regulating hemostasis and thrombosis. Rho GTPases are well known as molecular switches that control various cellular functions via a balanced GTP-binding/GTP-hydrolysis cycle and signaling cascade through downstream effectors. In platelets, Rho GTPases function as critical regulators by mediating signal transduction that drives platelet activation and aggregation. Mostly by gene targeting and pharmacological inhibition approaches, Rho GTPase family members RhoA, Rac1, and Cdc42 have been shown to be indispensable in regulating the actin cytoskeleton dynamics in platelets, affecting platelet shape change, spreading, secretion, and aggregation, leading to thrombus formation. Additionally, studies of Rho GTPase function using platelets as a non-transformed model due to their anucleated nature have revealed valuable information on cell signaling principles. This review provides an updated summary of recent advances in Rho GTPase signaling in platelet regulation. We also highlight pharmacological approaches that effectively inhibited platelet activation to explore their possible development into future antiplatelet therapies.
Collapse
Affiliation(s)
- Akhila Dandamudi
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
- Department of Pathology, University of Cincinnati Graduate School, Cincinnati, OH 45267, USA
| | - Huzoor Akbar
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | - Jose Cancelas
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
- Hoxworth Blood Center, College of Medicine, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Yi Zheng
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
- Department of Pathology, University of Cincinnati Graduate School, Cincinnati, OH 45267, USA
- Correspondence: ; Tel.: +1-513-636-0595
| |
Collapse
|
10
|
ROCK Inhibition as Potential Target for Treatment of Pulmonary Hypertension. Cells 2021; 10:cells10071648. [PMID: 34209333 PMCID: PMC8303917 DOI: 10.3390/cells10071648] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/17/2021] [Accepted: 06/23/2021] [Indexed: 02/07/2023] Open
Abstract
Pulmonary hypertension (PH) is a cardiovascular disease caused by extensive vascular remodeling in the lungs, which ultimately leads to death in consequence of right ventricle (RV) failure. While current drugs for PH therapy address the sustained vasoconstriction, no agent effectively targets vascular cell proliferation and tissue inflammation. Rho-associated protein kinases (ROCKs) emerged in the last few decades as promising targets for PH therapy, since ROCK inhibitors demonstrated significant anti-remodeling and anti-inflammatory effects. In this review, current aspects of ROCK inhibition therapy are discussed in relation to the treatment of PH and RV dysfunction, from cell biology to preclinical and clinical studies.
Collapse
|
11
|
Duan X, Perveen R, Dandamudi A, Adili R, Johnson J, Funk K, Berryman M, Davis AK, Holinstat M, Zheng Y, Akbar H. Pharmacologic targeting of Cdc42 GTPase by a small molecule Cdc42 activity-specific inhibitor prevents platelet activation and thrombosis. Sci Rep 2021; 11:13170. [PMID: 34162972 PMCID: PMC8222210 DOI: 10.1038/s41598-021-92654-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 05/27/2021] [Indexed: 01/14/2023] Open
Abstract
Gene targeting of Cdc42 GTPase has been shown to inhibit platelet activation. In this study, we investigated a hypothesis that inhibition of Cdc42 activity by CASIN, a small molecule Cdc42 Activity-Specific INhibitor, may down regulate platelet activation and thrombus formation. We investigated the effects of CASIN on platelet activation in vitro and thrombosis in vivo. In human platelets, CASIN, but not its inactive analog Pirl7, blocked collagen induced activation of Cdc42 and inhibited phosphorylation of its downstream effector, PAK1/2. Moreover, addition of CASIN to washed human platelets inhibited platelet spreading on immobilized fibrinogen. Treatment of human platelets with CASIN inhibited collagen or thrombin induced: (a) ATP secretion and platelet aggregation; and (b) phosphorylation of Akt, ERK and p38-MAPK. Pre-incubation of platelets with Pirl7, an inactive analog of CASIN, failed to inhibit collagen induced aggregation. Washing of human platelets after incubation with CASIN eliminated its inhibitory effect on collagen induced aggregation. Intraperitoneal administration of CASIN to wild type mice inhibited ex vivo aggregation induced by collagen but did not affect the murine tail bleeding times. CASIN administration, prior to laser-induced injury in murine cremaster muscle arterioles, resulted in formation of smaller and unstable thrombi compared to control mice without CASIN treatment. These data suggest that pharmacologic targeting of Cdc42 by specific and reversible inhibitors may lead to the discovery of novel antithrombotic agents.
Collapse
Affiliation(s)
- Xin Duan
- Division of Experimental Hematology and Cancer Biology, Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, 45229, USA
| | - Rehana Perveen
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, 45701, USA
| | - Akhila Dandamudi
- Division of Experimental Hematology and Cancer Biology, Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, 45229, USA
| | - Reheman Adili
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - James Johnson
- Division of Experimental Hematology and Cancer Biology, Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, 45229, USA
| | - Kevin Funk
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, 45701, USA
| | - Mark Berryman
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, 45701, USA
| | - Ashley Kuenzi Davis
- Division of Experimental Hematology and Cancer Biology, Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, 45229, USA
| | - Michael Holinstat
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Yi Zheng
- Division of Experimental Hematology and Cancer Biology, Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, 45229, USA.
| | - Huzoor Akbar
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, 45701, USA.
| |
Collapse
|
12
|
Xu Z, Liang Y, Delaney MK, Zhang Y, Kim K, Li J, Bai Y, Cho J, Ushio-Fukai M, Cheng N, Du X. Shear and Integrin Outside-In Signaling Activate NADPH-Oxidase 2 to Promote Platelet Activation. Arterioscler Thromb Vasc Biol 2021; 41:1638-1653. [PMID: 33691478 PMCID: PMC8057529 DOI: 10.1161/atvbaha.120.315773] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 03/01/2021] [Indexed: 11/18/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Zheng Xu
- Department of Pharmacology, University of Illinois at Chicago (Z.X., Y.L., M.K.D., Y.Z., K.K., J.L., Y.B., J.C., M.U.-F., N.C., X.D.)
| | - Ying Liang
- Department of Pharmacology, University of Illinois at Chicago (Z.X., Y.L., M.K.D., Y.Z., K.K., J.L., Y.B., J.C., M.U.-F., N.C., X.D.)
| | - M. Keegan Delaney
- Department of Pharmacology, University of Illinois at Chicago (Z.X., Y.L., M.K.D., Y.Z., K.K., J.L., Y.B., J.C., M.U.-F., N.C., X.D.)
- Dupage Medical Technology, Inc (M.K.D.)
| | - Yaping Zhang
- Department of Pharmacology, University of Illinois at Chicago (Z.X., Y.L., M.K.D., Y.Z., K.K., J.L., Y.B., J.C., M.U.-F., N.C., X.D.)
| | - Kyungho Kim
- Department of Pharmacology, University of Illinois at Chicago (Z.X., Y.L., M.K.D., Y.Z., K.K., J.L., Y.B., J.C., M.U.-F., N.C., X.D.)
- Korean Medicine-Application Center, Korea Institute of Oriental Medicine, Daegu (K.K.)
| | - Jing Li
- Department of Pharmacology, University of Illinois at Chicago (Z.X., Y.L., M.K.D., Y.Z., K.K., J.L., Y.B., J.C., M.U.-F., N.C., X.D.)
| | - Yanyan Bai
- Department of Pharmacology, University of Illinois at Chicago (Z.X., Y.L., M.K.D., Y.Z., K.K., J.L., Y.B., J.C., M.U.-F., N.C., X.D.)
| | - Jaehyung Cho
- Department of Pharmacology, University of Illinois at Chicago (Z.X., Y.L., M.K.D., Y.Z., K.K., J.L., Y.B., J.C., M.U.-F., N.C., X.D.)
| | - Masuko Ushio-Fukai
- Department of Pharmacology, University of Illinois at Chicago (Z.X., Y.L., M.K.D., Y.Z., K.K., J.L., Y.B., J.C., M.U.-F., N.C., X.D.)
- Department of Medicine (Cardiology), Vascular Biology Center, Medical College of Georgia at Augusta University (M.U.-F.)
| | - Ni Cheng
- Department of Pharmacology, University of Illinois at Chicago (Z.X., Y.L., M.K.D., Y.Z., K.K., J.L., Y.B., J.C., M.U.-F., N.C., X.D.)
| | - Xiaoping Du
- Department of Pharmacology, University of Illinois at Chicago (Z.X., Y.L., M.K.D., Y.Z., K.K., J.L., Y.B., J.C., M.U.-F., N.C., X.D.)
| |
Collapse
|
13
|
Ngo ATP, Parra-Izquierdo I, Aslan JE, McCarty OJT. Rho GTPase regulation of reactive oxygen species generation and signalling in platelet function and disease. Small GTPases 2021; 12:440-457. [PMID: 33459160 DOI: 10.1080/21541248.2021.1878001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
Platelets are master regulators and effectors of haemostasis with increasingly recognized functions as mediators of inflammation and immune responses. The Rho family of GTPase members Rac1, Cdc42 and RhoA are known to be major components of the intracellular signalling network critical to platelet shape change and morphological dynamics, thus playing a major role in platelet spreading, secretion and thrombus formation. Initially linked to the regulation of actomyosin contraction and lamellipodia formation, recent reports have uncovered non-canonical functions of platelet RhoGTPases in the regulation of reactive oxygen species (ROS), where intrinsically generated ROS modulate platelet function and contribute to thrombus formation. Platelet RhoGTPases orchestrate oxidative processes and cytoskeletal rearrangement in an interconnected manner to regulate intracellular signalling networks underlying platelet activity and thrombus formation. Herein we review our current knowledge of the regulation of platelet ROS generation by RhoGTPases and their relationship with platelet cytoskeletal reorganization, activation and function.
Collapse
Affiliation(s)
- Anh T P Ngo
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA
| | - Ivan Parra-Izquierdo
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA.,Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Joseph E Aslan
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA.,Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon, USA.,Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, Oregon, USA
| | - Owen J T McCarty
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
14
|
Cioffi F, Adam RHI, Broersen K. Molecular Mechanisms and Genetics of Oxidative Stress in Alzheimer's Disease. J Alzheimers Dis 2020; 72:981-1017. [PMID: 31744008 PMCID: PMC6971833 DOI: 10.3233/jad-190863] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Alzheimer’s disease is the most common neurodegenerative disorder that can cause dementia in elderly over 60 years of age. One of the disease hallmarks is oxidative stress which interconnects with other processes such as amyloid-β deposition, tau hyperphosphorylation, and tangle formation. This review discusses current thoughts on molecular mechanisms that may relate oxidative stress to Alzheimer’s disease and identifies genetic factors observed from in vitro, in vivo, and clinical studies that may be associated with Alzheimer’s disease-related oxidative stress.
Collapse
Affiliation(s)
- Federica Cioffi
- Nanobiophysics Group, Technical Medical Centre, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands
| | - Rayan Hassan Ibrahim Adam
- Nanobiophysics Group, Technical Medical Centre, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands
| | - Kerensa Broersen
- Applied Stem Cell Technologies, Technical Medical Centre, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands
| |
Collapse
|
15
|
Hegde S, Wellendorf AM, Zheng Y, Cancelas JA. Antioxidant prevents clearance of hemostatically competent platelets after long-term cold storage. Transfusion 2020; 61:557-567. [PMID: 33247486 DOI: 10.1111/trf.16200] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 09/27/2020] [Accepted: 10/23/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUND Cold storage of platelets (PLTs) has the potential advantage of prolonging storage time while reducing posttransfusion infection given the decreased likelihood of bacterial outgrowth during storage and possibly beneficial effects in treating bleeding patients. However, cold storage reduces PLT survival through the induction of complex storage lesions, which are more accentuated when storage is prolonged. STUDY DESIGN AND METHODS Whole blood-derived PLT-rich plasma concentrates from seven PLT pools (n = 5 donors per pool). PLT additive solution was added (67%/33% plasma) and the product was split into 50-mL bags. Split units were stored in the presence or absence of 1 mM of N-acetylcysteine (NAC) under agitation for up to 14 days at room temperature or in the cold and were analyzed for PLT activation, fibrinogen-dependent spreading, microparticle formation, mitochondrial respiratory activity, reactive oxygen species (ROS) generation, as well as in vivo survival and bleeding time correction in immunodeficient mice. RESULTS Cold storage of PLTs for 7 days or longer induces significant PLT activation, cytoskeletal damage, impaired fibrinogen spreading, enhances mitochondrial metabolic decoupling and ROS generation, and increases macrophage-dependent phagocytosis and macrophage-independent clearance. Addition of NAC prevents PLT clearance and allows a correction of the prolonged bleeding time in thrombocytopenic, aspirin-treated, immunodeficient mice. CONCLUSIONS Long-term cold storage induces mitochondrial uncoupling and increased proton leak and ROS generation. The resulting ROS is a crucial contributor to the increased macrophage-dependent and -independent clearance of functional PLTs and can be prevented by the antioxidant NAC in a magnesium-containing additive solution.
Collapse
Affiliation(s)
- Shailaja Hegde
- Hoxworth Blood Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Ashley M Wellendorf
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Yi Zheng
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Jose A Cancelas
- Hoxworth Blood Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
16
|
Small G-protein RhoA is a potential inhibitor of cardiac fast sodium current. J Physiol Biochem 2020; 77:13-23. [PMID: 33145656 DOI: 10.1007/s13105-020-00774-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 10/27/2020] [Indexed: 12/30/2022]
Abstract
Small G-proteins of Rho family modulate the activity of several classes of ion channels, including K+ channels Kv1.2, Kir2.1, and ERG; Ca2+ channels; and epithelial Na+ channels. The present study was aimed to check the RhoA potential regulatory effects on Na+ current (INa) transferred by Na+ channel cardiac isoform NaV1.5 in heterologous expression system and in native rat cardiomyocytes. Whole-cell patch-clamp experiments showed that coexpression of NaV1.5 with the wild-type RhoA in CHO-K1 cell line caused 2.7-fold decrease of INa density with minimal influence on steady-state activation and inactivation. This effect was reproduced by the coexpression with a constitutively active RhoA, but not with a dominant negative RhoA. In isolated ventricular rat cardiomyocytes, a 5-h incubation with the RhoA activator narciclasine (5 × 10-6 M) reduced the maximal INa density by 38.8%. The RhoA-selective inhibitor rhosin (10-5 M) increased the maximal INa density by 25.3%. Experiments with sharp microelectrode recordings in isolated right ventricular wall preparations showed that 5 × 10-6 M narciclasine induced a significant reduction of action potential upstroke velocity after 2 h of incubation. Thus, RhoA might be considered as a potential negative regulator of sodium channels cardiac isoform NaV1.5.
Collapse
|
17
|
Vara D, Tarafdar A, Celikag M, Patinha D, Gulacsy CE, Hounslea E, Warren Z, Ferreira B, Koeners MP, Caggiano L, Pula G. NADPH oxidase 1 is a novel pharmacological target for the development of an antiplatelet drug without bleeding side effects. FASEB J 2020; 34:13959-13977. [PMID: 32851720 DOI: 10.1096/fj.202001086rrr] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 08/01/2020] [Accepted: 08/07/2020] [Indexed: 12/25/2022]
Abstract
Growing evidence supports a central role of NADPH oxidases (NOXs) in the regulation of platelets, which are circulating cells involved in both hemostasis and thrombosis. Here, the use of Nox1-/- and Nox1+/+ mice as experimental models of human responses demonstrated a critical role of NOX1 in collagen-dependent platelet activation and pathological arterial thrombosis, as tested in vivo by carotid occlusion assays. In contrast, NOX1 does not affect platelet responses to thrombin and normal hemostasis, as assayed in tail bleeding experiments. Therefore, as NOX1 inhibitors are likely to have antiplatelet effects without associated bleeding risks, the NOX1-selective inhibitor 2-acetylphenothiazine (2APT) and a series of its derivatives generated to increase inhibitory potency and drug bioavailability were tested. Among the 2APT derivatives, 1-(10H-phenothiazin-2-yl)vinyl tert-butyl carbonate (2APT-D6) was selected for its high potency. Both 2APT and 2APT-D6 inhibited collagen-dependent platelet aggregation, adhesion, thrombus formation, superoxide anion generation, and surface activation marker expression, while responses to thrombin or adhesion to fibrinogen were not affected. In vivo administration of 2APT or 2APT-D6 led to the inhibition of mouse platelet aggregation, oxygen radical output, and thrombus formation, and carotid occlusion, while tail hemostasis was unaffected. Differently to in vitro experiments, 2APT-D6 and 2APT displayed similar potency in vivo. In summary, NOX1 inhibition with 2APT or its derivative 2APT-D6 is a viable strategy to control collagen-induced platelet activation and reduce thrombosis without deleterious effects on hemostasis. These compounds should, therefore, be considered for the development of novel antiplatelet drugs to fight cardiovascular diseases in humans.
Collapse
Affiliation(s)
- Dina Vara
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, UK
| | - Anuradha Tarafdar
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, UK
| | - Meral Celikag
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, UK
| | - Daniela Patinha
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, UK
| | | | - Ellie Hounslea
- Department of Pharmacy and Pharmacology, University of Bath, Bath, UK
| | - Zach Warren
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, UK
| | - Barbara Ferreira
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, UK
| | - Maarten P Koeners
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, UK
| | - Lorenzo Caggiano
- Department of Pharmacy and Pharmacology, University of Bath, Bath, UK
| | - Giordano Pula
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
18
|
The choline transporter Slc44a2 controls platelet activation and thrombosis by regulating mitochondrial function. Nat Commun 2020; 11:3479. [PMID: 32661250 PMCID: PMC7359028 DOI: 10.1038/s41467-020-17254-w] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 06/15/2020] [Indexed: 02/08/2023] Open
Abstract
Genetic factors contribute to the risk of thrombotic diseases. Recent genome wide association studies have identified genetic loci including SLC44A2 which may regulate thrombosis. Here we show that Slc44a2 controls platelet activation and thrombosis by regulating mitochondrial energetics. We find that Slc44a2 null mice (Slc44a2(KO)) have increased bleeding times and delayed thrombosis compared to wild-type (Slc44a2(WT)) controls. Platelets from Slc44a2(KO) mice have impaired activation in response to thrombin. We discover that Slc44a2 mediates choline transport into mitochondria, where choline metabolism leads to an increase in mitochondrial oxygen consumption and ATP production. Platelets lacking Slc44a2 contain less ATP at rest, release less ATP when activated, and have an activation defect that can be rescued by exogenous ADP. Taken together, our data suggest that mitochondria require choline for maximum function, demonstrate the importance of mitochondrial metabolism to platelet activation, and reveal a mechanism by which Slc44a2 influences thrombosis. Genetic association studies have identified loci including the choline transporter SLC44A2 as a potential regulator of thrombosis. Here the authors report that loss of SLC44A2 impairs platelet activation and thrombosis in mice via a reduction of mitochondrial ATP production.
Collapse
|
19
|
Sema3A - mediated modulation of NR1D1 expression may be involved in the regulation of axonal guidance signaling by the microbiota. Life Sci 2019; 223:54-61. [DOI: 10.1016/j.lfs.2019.03.023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 03/02/2019] [Accepted: 03/09/2019] [Indexed: 01/13/2023]
|
20
|
Truong NF, Lesher-Pérez SC, Kurt E, Segura T. Pathways Governing Polyethylenimine Polyplex Transfection in Microporous Annealed Particle Scaffolds. Bioconjug Chem 2019; 30:476-486. [PMID: 30513197 PMCID: PMC7290906 DOI: 10.1021/acs.bioconjchem.8b00696] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Gene delivery using injectable hydrogels can serve as a potential method for regulated tissue regeneration in wound healing. Our microporous annealed particle (MAP) hydrogel has been shown to promote cellular infiltration in both skin and brain wounds, while reducing inflammation. Although the scaffold itself can promote healing, it is likely that other signals will be required to promote healing of hard-to-treat wounds. Gene delivery is one approach to introduce desired bioactive signals. In this study, we investigated how the properties of MAP hydrogels influence non-viral gene delivery of polyethylenimine-condensed plasmid to cells seeded within the MAP gel. From past studies, we found that gene transfer to cells seeded in tissue culture plastic differed from gene transfer to cells seeded inside hydrogel scaffolds. Since MAP scaffolds are generated from hydrogel microparticles that are approximately 100 μm in diameter, they display local characteristics that can be viewed as two-dimensional or three-dimensional to cells. Thus, we sought to study if gene transfer inside MAP scaffolds differed from gene transfer to cells seeded in tissue culture plastic. We sought to understand the roles of the endocytosis pathway, actin and microtubule dynamics, RhoGTPases, and YAP/TAZ on transfection of human fibroblasts.
Collapse
Affiliation(s)
- Norman F Truong
- Department of Chemical and Biomolecular Engineering , University of California , Los Angeles , California 90095 , United States
| | - Sasha Cai Lesher-Pérez
- Department of Chemical and Biomolecular Engineering , University of California , Los Angeles , California 90095 , United States
| | - Evan Kurt
- Department of Biomedical Engineering , Duke University , Durham , North Carolina 27708 , United States
| | - Tatiana Segura
- Department of Chemical and Biomolecular Engineering , University of California , Los Angeles , California 90095 , United States
| |
Collapse
|
21
|
Abstract
SIGNIFICANCE G protein-coupled receptors (GPCR) are the largest group of cell surface receptors, which link cells to their environment. Reactive oxygen species (ROS) can act as important cellular signaling molecules. The family of NADPH oxidases generates ROS in response to activated cell surface receptors. Recent Advances: Various signaling pathways linking GPCRs and activation of NADPH oxidases have been characterized. CRITICAL ISSUES Still, a more detailed analysis of G proteins involved in the GPCR-mediated activation of NADPH oxidases is needed. In addition, a more precise discrimination of NADPH oxidase activation due to either upregulation of subunit expression or post-translational subunit modifications is needed. Also, the role of noncanonical modulators of NADPH oxidase activation in the response to GPCRs awaits further analyses. FUTURE DIRECTIONS As GPCRs are one of the most popular classes of investigational drug targets, further detailing of G protein-coupled mechanisms in the activation mechanism of NADPH oxidases as well as better understanding of the link between newly identified NADPH oxidase interaction partners and GPCR signaling will provide new opportunities for improved efficiency and decreased off target effects of therapies targeting GPCRs.
Collapse
Affiliation(s)
- Andreas Petry
- 1 Experimental and Molecular Pediatric Cardiology, German Heart Center Munich , TU Munich, Munich, Germany
| | - Agnes Görlach
- 1 Experimental and Molecular Pediatric Cardiology, German Heart Center Munich , TU Munich, Munich, Germany .,2 DZHK (German Centre for Cardiovascular Research) , Partner Site Munich, Munich Heart Alliance, Munich, Germany
| |
Collapse
|
22
|
NADPH oxidase 2 (NOX2): A key target of oxidative stress-mediated platelet activation and thrombosis. Trends Cardiovasc Med 2018; 28:429-434. [DOI: 10.1016/j.tcm.2018.03.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 02/24/2018] [Accepted: 03/09/2018] [Indexed: 01/01/2023]
|
23
|
Akbar H, Duan X, Piatt R, Saleem S, Davis AK, Tandon NN, Bergmeier W, Zheng Y. Small molecule targeting the Rac1-NOX2 interaction prevents collagen-related peptide and thrombin-induced reactive oxygen species generation and platelet activation. J Thromb Haemost 2018; 16:2083-2096. [PMID: 30007118 PMCID: PMC6472274 DOI: 10.1111/jth.14240] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Indexed: 12/29/2022]
Abstract
Essentials Reactive oxygen species (ROS) generation by NOX2 plays a critical role in platelet activation. Rac1 regulation of NOX2 is important for ROS generation. Small molecule inhibitor of the Rac1-p67phox interaction prevents platelet activation. Pharmacologic targeting of Rac1-NOX2 axis can be a viable approach for antithrombotic therapy. SUMMARY Background Platelets from patients with X-linked chronic granulomatous disease or mice deficient in nicotinamide adenine dinucleotide (phosphate) (NAD(P)H) oxidase isoform NOX2 exhibit diminished reactive oxygen species (ROS) generation and platelet activation. Binding of Rac1 GTPase to p67phox plays a critical role in NOX2 activation by facilitating the assembly of the NOX2 enzyme complex. Objective We tested the hypothesis that Phox-I, a rationally designed small molecule inhibitor of Rac-p67phox interaction, may serve as an antithrombosis agent by suppressing ROS production and platelet activation. Results Collagen-related peptide (CRP) induced ROS generation in a time-dependent manner. Platelets from Rac1-/- mice or human platelets treated with NSC23766, a specific Rac inhibitor, produced significantly less ROS in response to CRP. Treatment of platelets with Phox-I inhibited diverse CRP-induced responses, including: (i) ROS generation; (ii) release of P-selectin; (iii) secretion of ATP; (iv) platelet aggregation; and (v) phosphorylation of Akt. Similarly, incubation of platelets with Phox-I inhibited thrombin-induced: (i) secretion of ATP; (ii) platelet aggregation; (iii) rise in cytosolic calcium; and (iv) phosphorylation of Akt. In mouse models, intraperitoneal administration of Phox-I inhibited: (i) collagen-induced platelet aggregation without affecting the tail bleeding time and (ii) in vivo platelet adhesion/accumulation at the laser injury sites on the saphenous vein without affecting the time for complete cessation of blood loss. Conclusions Small molecule targeting of the Rac1-p67phox interaction may present an antithrombosis regimen by preventing GPVI- and non-GPVI-mediated NOX2 activation, ROS generation and platelet function without affecting the bleeding time.
Collapse
Affiliation(s)
- H Akbar
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - X Duan
- Division of Experimental Hematology and Cancer Biology, Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, USA
| | - R Piatt
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA
| | - S Saleem
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - A K Davis
- Division of Experimental Hematology and Cancer Biology, Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, USA
| | | | - W Bergmeier
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA
| | - Y Zheng
- Division of Experimental Hematology and Cancer Biology, Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
24
|
Affiliation(s)
- Joseph E. Aslan
- Knight Cardiovascular Institute, School of Medicine, Oregon Health & Science University, Portland, Oregon, USA
- Department of Biochemistry and Molecular Biology, School of Medicine, Oregon Health & Science University, Portland, Oregon, USA
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
25
|
Cameron SJ, Mix DS, Ture SK, Schmidt RA, Mohan A, Pariser D, Stoner MC, Shah P, Chen L, Zhang H, Field DJ, Modjeski KL, Toth S, Morrell CN. Hypoxia and Ischemia Promote a Maladaptive Platelet Phenotype. Arterioscler Thromb Vasc Biol 2018; 38:1594-1606. [PMID: 29724818 PMCID: PMC6023774 DOI: 10.1161/atvbaha.118.311186] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 04/17/2018] [Indexed: 12/26/2022]
Abstract
Supplemental Digital Content is available in the text. Objective— Reduced blood flow and tissue oxygen tension conditions result from thrombotic and vascular diseases such as myocardial infarction, stroke, and peripheral vascular disease. It is largely assumed that while platelet activation is increased by an acute vascular event, chronic vascular inflammation, and ischemia, the platelet activation pathways and responses are not themselves changed by the disease process. We, therefore, sought to determine whether the platelet phenotype is altered by hypoxic and ischemic conditions. Approach and Results— In a cohort of patients with metabolic and peripheral artery disease, platelet activity was enhanced, and inhibition with oral antiplatelet agents was impaired compared with platelets from control subjects, suggesting a difference in platelet phenotype caused by the disease. Isolated murine and human platelets exposed to reduced oxygen (hypoxia chamber, 5% O2) had increased expression of some proteins that augment platelet activation compared with platelets in normoxic conditions (21% O2). Using a murine model of critical limb ischemia, platelet activity was increased even 2 weeks postsurgery compared with sham surgery mice. This effect was partly inhibited in platelet-specific ERK5 (extracellular regulated protein kinase 5) knockout mice. Conclusions— These findings suggest that ischemic disease changes the platelet phenotype and alters platelet agonist responses because of changes in the expression of signal transduction pathway proteins. Platelet phenotype and function should, therefore, be better characterized in ischemic and hypoxic diseases to understand the benefits and limitations of antiplatelet therapy.
Collapse
Affiliation(s)
- Scott J Cameron
- From the Aab Cardiovascular Research Institute (S.J.C., S.K.T., R.A.S., A.M., D.P., D.J.F., K.L.M., C.N.M.) .,Division of Cardiology, Department of Medicine (S.J.C., C.N.M.)
| | - Doran S Mix
- Division of Vascular Surgery, Department of Surgery (D.S.M., M.C.S., S.T.), University of Rochester School of Medicine, NY
| | - Sara K Ture
- From the Aab Cardiovascular Research Institute (S.J.C., S.K.T., R.A.S., A.M., D.P., D.J.F., K.L.M., C.N.M.)
| | - Rachel A Schmidt
- From the Aab Cardiovascular Research Institute (S.J.C., S.K.T., R.A.S., A.M., D.P., D.J.F., K.L.M., C.N.M.)
| | - Amy Mohan
- From the Aab Cardiovascular Research Institute (S.J.C., S.K.T., R.A.S., A.M., D.P., D.J.F., K.L.M., C.N.M.)
| | - Daphne Pariser
- From the Aab Cardiovascular Research Institute (S.J.C., S.K.T., R.A.S., A.M., D.P., D.J.F., K.L.M., C.N.M.)
| | - Michael C Stoner
- Division of Vascular Surgery, Department of Surgery (D.S.M., M.C.S., S.T.), University of Rochester School of Medicine, NY
| | - Punit Shah
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD (P.S., L.C., H.Z.)
| | - Lijun Chen
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD (P.S., L.C., H.Z.)
| | - Hui Zhang
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD (P.S., L.C., H.Z.)
| | - David J Field
- From the Aab Cardiovascular Research Institute (S.J.C., S.K.T., R.A.S., A.M., D.P., D.J.F., K.L.M., C.N.M.)
| | - Kristina L Modjeski
- From the Aab Cardiovascular Research Institute (S.J.C., S.K.T., R.A.S., A.M., D.P., D.J.F., K.L.M., C.N.M.)
| | - Sandra Toth
- Division of Vascular Surgery, Department of Surgery (D.S.M., M.C.S., S.T.), University of Rochester School of Medicine, NY
| | - Craig N Morrell
- From the Aab Cardiovascular Research Institute (S.J.C., S.K.T., R.A.S., A.M., D.P., D.J.F., K.L.M., C.N.M.).,Division of Cardiology, Department of Medicine (S.J.C., C.N.M.)
| |
Collapse
|
26
|
Wang Y, Jiang L, Mo X, Lan Y, Yang X, Liu X, Zhang J, Zhu L, Liu J, Wu X. Megakaryocytic Smad4 Regulates Platelet Function through Syk and ROCK2 Expression. Mol Pharmacol 2017; 92:285-296. [PMID: 28663280 DOI: 10.1124/mol.116.107417] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 06/21/2017] [Indexed: 01/03/2023] Open
Abstract
Smad4, a key transcription factor in the transforming growth factor-β signaling pathway, is involved in a variety of cell physiologic and pathologic processes. Here, we characterized megakaryocyte/platelet-specific Smad4 deficiency in mice to elucidate its effect on platelet function. We found that megakaryocyte/platelet-specific loss of Smad4 caused mild thrombocytopenia and significantly extended first occlusion time and tail bleeding time in mice. Smad4-deficient platelets showed reduced agonist-induced platelet aggregation. Further studies showed that a severe defect was seen in integrin αIIbβ3-mediated bidirectional (inside-out and outside-in) signaling in Smad4-deficient platelets, as evidenced by reduced fibrinogen binding and α-granule secretion, suppressed platelet spreading and clot retraction. Microarray analysis showed that the expression levels of multiple genes were altered in Smad4-deficient platelets. Among these genes, spleen tyrosine kinase (Syk) and Rho-associated coiled-coil containing protein kinase 2 (ROCK2) were downregulated several times as confirmed by quantitative reverse-transcription polymerase chain reaction and immunoblotting. Further research showed that Smad4 directly regulates ROCK2 transcription but indirectly regulates Syk. Megakaryocyte/platelet-specific Smad4 deficiency caused decreased expression levels of Syk and ROCK2 in platelets. These results suggest potential links among Smad4 deficiency, attenuated Syk, and ROCK2 expression and defective platelet activation.
Collapse
Affiliation(s)
- Yanhua Wang
- Department of Laboratory Medicine, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, People's Republic of China (Y.W.); Institute for Pediatric Translational Medicine, Shanghai Children's Medical Center, Shanghai, People's Republic of China (L.J., X.M.); State Key Laboratory of Proteomics, Genetic Laboratory of Development and Diseases, Institute of Biotechnology, Beijing, People's Republic of China (Y.L., X.Y.); Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China (X.L., J.Z.); Cyrus Tang Hematology Center, Soochow University, Suzhou, People's Republic of China (L.Z.); Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China (J.L.); and The Central Laboratory of The Eighth People's Hospital of Shanghai, Shanghai, People's Republic of China (X.W.)
| | - Lirong Jiang
- Department of Laboratory Medicine, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, People's Republic of China (Y.W.); Institute for Pediatric Translational Medicine, Shanghai Children's Medical Center, Shanghai, People's Republic of China (L.J., X.M.); State Key Laboratory of Proteomics, Genetic Laboratory of Development and Diseases, Institute of Biotechnology, Beijing, People's Republic of China (Y.L., X.Y.); Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China (X.L., J.Z.); Cyrus Tang Hematology Center, Soochow University, Suzhou, People's Republic of China (L.Z.); Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China (J.L.); and The Central Laboratory of The Eighth People's Hospital of Shanghai, Shanghai, People's Republic of China (X.W.)
| | - Xi Mo
- Department of Laboratory Medicine, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, People's Republic of China (Y.W.); Institute for Pediatric Translational Medicine, Shanghai Children's Medical Center, Shanghai, People's Republic of China (L.J., X.M.); State Key Laboratory of Proteomics, Genetic Laboratory of Development and Diseases, Institute of Biotechnology, Beijing, People's Republic of China (Y.L., X.Y.); Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China (X.L., J.Z.); Cyrus Tang Hematology Center, Soochow University, Suzhou, People's Republic of China (L.Z.); Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China (J.L.); and The Central Laboratory of The Eighth People's Hospital of Shanghai, Shanghai, People's Republic of China (X.W.)
| | - Yu Lan
- Department of Laboratory Medicine, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, People's Republic of China (Y.W.); Institute for Pediatric Translational Medicine, Shanghai Children's Medical Center, Shanghai, People's Republic of China (L.J., X.M.); State Key Laboratory of Proteomics, Genetic Laboratory of Development and Diseases, Institute of Biotechnology, Beijing, People's Republic of China (Y.L., X.Y.); Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China (X.L., J.Z.); Cyrus Tang Hematology Center, Soochow University, Suzhou, People's Republic of China (L.Z.); Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China (J.L.); and The Central Laboratory of The Eighth People's Hospital of Shanghai, Shanghai, People's Republic of China (X.W.)
| | - Xiao Yang
- Department of Laboratory Medicine, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, People's Republic of China (Y.W.); Institute for Pediatric Translational Medicine, Shanghai Children's Medical Center, Shanghai, People's Republic of China (L.J., X.M.); State Key Laboratory of Proteomics, Genetic Laboratory of Development and Diseases, Institute of Biotechnology, Beijing, People's Republic of China (Y.L., X.Y.); Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China (X.L., J.Z.); Cyrus Tang Hematology Center, Soochow University, Suzhou, People's Republic of China (L.Z.); Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China (J.L.); and The Central Laboratory of The Eighth People's Hospital of Shanghai, Shanghai, People's Republic of China (X.W.)
| | - Xinyi Liu
- Department of Laboratory Medicine, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, People's Republic of China (Y.W.); Institute for Pediatric Translational Medicine, Shanghai Children's Medical Center, Shanghai, People's Republic of China (L.J., X.M.); State Key Laboratory of Proteomics, Genetic Laboratory of Development and Diseases, Institute of Biotechnology, Beijing, People's Republic of China (Y.L., X.Y.); Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China (X.L., J.Z.); Cyrus Tang Hematology Center, Soochow University, Suzhou, People's Republic of China (L.Z.); Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China (J.L.); and The Central Laboratory of The Eighth People's Hospital of Shanghai, Shanghai, People's Republic of China (X.W.)
| | - Jian Zhang
- Department of Laboratory Medicine, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, People's Republic of China (Y.W.); Institute for Pediatric Translational Medicine, Shanghai Children's Medical Center, Shanghai, People's Republic of China (L.J., X.M.); State Key Laboratory of Proteomics, Genetic Laboratory of Development and Diseases, Institute of Biotechnology, Beijing, People's Republic of China (Y.L., X.Y.); Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China (X.L., J.Z.); Cyrus Tang Hematology Center, Soochow University, Suzhou, People's Republic of China (L.Z.); Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China (J.L.); and The Central Laboratory of The Eighth People's Hospital of Shanghai, Shanghai, People's Republic of China (X.W.)
| | - Li Zhu
- Department of Laboratory Medicine, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, People's Republic of China (Y.W.); Institute for Pediatric Translational Medicine, Shanghai Children's Medical Center, Shanghai, People's Republic of China (L.J., X.M.); State Key Laboratory of Proteomics, Genetic Laboratory of Development and Diseases, Institute of Biotechnology, Beijing, People's Republic of China (Y.L., X.Y.); Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China (X.L., J.Z.); Cyrus Tang Hematology Center, Soochow University, Suzhou, People's Republic of China (L.Z.); Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China (J.L.); and The Central Laboratory of The Eighth People's Hospital of Shanghai, Shanghai, People's Republic of China (X.W.)
| | - Junling Liu
- Department of Laboratory Medicine, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, People's Republic of China (Y.W.); Institute for Pediatric Translational Medicine, Shanghai Children's Medical Center, Shanghai, People's Republic of China (L.J., X.M.); State Key Laboratory of Proteomics, Genetic Laboratory of Development and Diseases, Institute of Biotechnology, Beijing, People's Republic of China (Y.L., X.Y.); Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China (X.L., J.Z.); Cyrus Tang Hematology Center, Soochow University, Suzhou, People's Republic of China (L.Z.); Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China (J.L.); and The Central Laboratory of The Eighth People's Hospital of Shanghai, Shanghai, People's Republic of China (X.W.)
| | - Xiaolin Wu
- Department of Laboratory Medicine, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, People's Republic of China (Y.W.); Institute for Pediatric Translational Medicine, Shanghai Children's Medical Center, Shanghai, People's Republic of China (L.J., X.M.); State Key Laboratory of Proteomics, Genetic Laboratory of Development and Diseases, Institute of Biotechnology, Beijing, People's Republic of China (Y.L., X.Y.); Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China (X.L., J.Z.); Cyrus Tang Hematology Center, Soochow University, Suzhou, People's Republic of China (L.Z.); Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China (J.L.); and The Central Laboratory of The Eighth People's Hospital of Shanghai, Shanghai, People's Republic of China (X.W.)
| |
Collapse
|
27
|
Wang XQ, Zhang YP, Zhang LM, Feng NN, Zhang MZ, Zhao ZG, Niu CY. Resveratrol enhances vascular reactivity in mice following lipopolysaccharide challenge via the RhoA-ROCK-MLCP pathway. Exp Ther Med 2017; 14:308-316. [PMID: 28672931 PMCID: PMC5488661 DOI: 10.3892/etm.2017.4486] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 03/31/2017] [Indexed: 12/12/2022] Open
Abstract
The aim of the present study was to identify whether sepsis-induced vascular hyporeactivity is associated with microcirculation disturbance and multiple organ injuries. The current study assessed the impact of resveratrol (Res) treatment on lipopolysaccharide (LPS) challenge mediated vascular hyporeactivity. Effects of Res treatment (30 mg/kg; i.m.) at 1 h following LPS stimulation (5 mg/kg; i.v.) on the survival time, mean arterial pressure (MAP), and maximal difference of MAP (ΔMAP) to norepinephrine (NE; 4.2 µg/kg) in mice were observed. The reactivity to gradient NE of isolated mesenteric arterioles and the association with the RhoA-RhoA kinase (ROCK)-myosin light chain phosphatase (MLCP) pathway were investigated by myography, and the signaling molecule protein levels were assessed using ELISA. Res treatment prolonged the survival time of mice subjected to LPS challenge, but did not prevent the LPS-induced hypotension and increase in ΔMAP. Res treatment and RhoA agonist U-46619 incubation prevented LPS-induced vascular hyporeactivity ex vivo, which were suppressed by incubation with ROCK inhibitor Y-27632. LPS-induced vascular hyporeactivity was not affected by the MLCP inhibitor okadaic acid incubation, but was further downregulated by the co-incubation of OA plus Y-27632. The inhibiting effect of Y-27632 on Res treatment was eradicated by incubation with U-46619. Furthermore, RhoA inhibitor C3 transferase did not significantly inhibit the enhancing role of Res treatment, which was further increased by U-46619 plus C3 transferase co-incubation. In addition, Res treatment eradicated the LPS-induced decreases in p-RhoA and p-Mypt1 levels and increases in MLCP levels. The results of the present study indicate that post-treatment of Res significantly ameliorates LPS-induced vascular hyporeactivity, which is associated with the activation of the RhoA-ROCK-MLCP pathway.
Collapse
Affiliation(s)
- Xu-Qing Wang
- Institute of Microcirculation, Hebei North University, Zhangjiakou, Hebei 075000, P.R. China
| | - Yu-Ping Zhang
- Institute of Microcirculation, Hebei North University, Zhangjiakou, Hebei 075000, P.R. China
| | - Li-Min Zhang
- Institute of Microcirculation, Hebei North University, Zhangjiakou, Hebei 075000, P.R. China
| | - Niu-Niu Feng
- Institute of Microcirculation, Hebei North University, Zhangjiakou, Hebei 075000, P.R. China
| | - Ming-Zhu Zhang
- Institute of Microcirculation, Hebei North University, Zhangjiakou, Hebei 075000, P.R. China
| | - Zi-Gang Zhao
- Institute of Microcirculation, Hebei North University, Zhangjiakou, Hebei 075000, P.R. China
| | - Chun-Yu Niu
- Institute of Microcirculation, Hebei North University, Zhangjiakou, Hebei 075000, P.R. China
| |
Collapse
|