1
|
Kim SY, Cheon J. Senescence-associated microvascular endothelial dysfunction: A focus on the blood-brain and blood-retinal barriers. Ageing Res Rev 2024; 100:102446. [PMID: 39111407 DOI: 10.1016/j.arr.2024.102446] [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: 03/08/2024] [Revised: 07/05/2024] [Accepted: 08/02/2024] [Indexed: 08/17/2024]
Abstract
The blood-brain barrier (BBB) and blood-retinal barrier (BRB) constitute critical physiochemical interfaces, precisely orchestrating the bidirectional communication between the brain/retina and blood. Increased permeability or leakage of these barriers has been demonstrably linked to age-related vascular and parenchymal damage. While it has been suggested that the gradual aging process may coincide with disruptions in these barriers, this phenomenon is significantly exacerbated in individuals with age-related neurodegenerative disorders (ARND). This review focuses on the microvascular endothelium, a key constituent of BBB and BRB, highlighting the impact of endothelial senescence on barrier dysfunction and exploring recent discoveries regarding core pathways implicated in its breakdown. Subsequently, we address the "vascular senescence hypothesis" for ARND, with a particular emphasis on Alzheimer's disease and age-related macular degeneration, centered on endothelial senescence. Finally, we discuss potential senotherapeutic strategies targeting barrier dysfunction.
Collapse
Affiliation(s)
- Sung Young Kim
- Department of Biochemistry, Konkuk University School of Medicine, Republic of Korea; Research Institute of Medical Science, Konkuk University, Republic of Korea; IBST, Konkuk University, Republic of Korea.
| | - Jaejoung Cheon
- Department of Biochemistry, Konkuk University School of Medicine, Republic of Korea
| |
Collapse
|
2
|
Zemskov EA, Zemskova MA, Wu X, Moreno Caceres S, Caraballo Delgado D, Yegambaram M, Lu Q, Fu P, Wang T, Black SM. Novel mechanism of cyclic nucleotide crosstalk mediated by PKG-dependent proteasomal degradation of the Hsp90 client protein phosphodiesterase 3A. J Biol Chem 2024; 300:107723. [PMID: 39214301 PMCID: PMC11497409 DOI: 10.1016/j.jbc.2024.107723] [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: 07/19/2023] [Revised: 08/04/2024] [Accepted: 08/13/2024] [Indexed: 09/04/2024] Open
Abstract
Endothelial cAMP-specific phosphodiesterase PDE3A is one of the major negative regulators of the endothelial barrier function in acute lung injury models. However, the mechanisms underlying its regulation still need to be fully resolved. We show here that the PDE3A is a newly described client of the molecular chaperone heat shock protein 90 (hsp90). In endothelial cells (ECs), hsp90 inhibition by geldanamycin (GA) led to a disruption of the hsp90/PDE3A complex, followed by a significant decrease in PDE3A protein levels. The decrease in PDE3A protein levels was ubiquitin-proteasome-dependent and required the activity of the E3 ubiquitin ligase C terminus of Hsc70-interacting protein. GA treatment also enhanced the association of PDE3A with hsp70, which partially prevented PDE3A degradation. GA-induced decreases in PDE3A protein levels correlated with decreased PDE3 activity and increased cAMP levels in EC. We also demonstrated that protein kinase G-dependent phosphorylation of PDE3A at Ser654 can signal the dissociation of PDE3A from hsp90 and PDE3A degradation. This was confirmed by endogenous PDE3A phosphorylation and degradation in 8-Br-cGMP- or 8-CPT-cGMP- and Bay 41-8543-stimulated EC and comparisons of WT- and phospho-mimic S654D mutant PDE3A protein stability in transiently transfected HEK293 cells. In conclusion, we have identified a new mechanism of PDE3A regulation mediated by the ubiquitin-proteasome system. Further, the degradation of PDE3A is controlled by the phosphorylation of S654 and the interaction with hsp90. We speculate that targeting the PDE3A/hsp90 complex could be a therapeutic approach for acute lung injury.
Collapse
Affiliation(s)
- Evgeny A Zemskov
- Center for Translational Science, Florida International University, Port St Lucie, Florida, USA; Cellular & Molecular Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida, USA
| | - Marina A Zemskova
- Center for Translational Science, Florida International University, Port St Lucie, Florida, USA
| | - Xiaomin Wu
- Department of Medicine, University of Arizona Health Sciences, Tucson, Arizona, USA
| | - Santiago Moreno Caceres
- Center for Translational Science, Florida International University, Port St Lucie, Florida, USA
| | - David Caraballo Delgado
- Center for Translational Science, Florida International University, Port St Lucie, Florida, USA
| | - Manivannan Yegambaram
- Center for Translational Science, Florida International University, Port St Lucie, Florida, USA
| | - Qing Lu
- Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Miami, Florida, USA
| | - Panfeng Fu
- Center for Translational Science, Florida International University, Port St Lucie, Florida, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Miami, Florida, USA
| | - Ting Wang
- Center for Translational Science, Florida International University, Port St Lucie, Florida, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Miami, Florida, USA
| | - Stephen M Black
- Center for Translational Science, Florida International University, Port St Lucie, Florida, USA; Cellular & Molecular Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Miami, Florida, USA.
| |
Collapse
|
3
|
Pokharel MD, Fu P, Garcia-Flores A, Yegambaram M, Lu Q, Sun X, Unwalla H, Aggarwal S, Fineman JR, Wang T, Black SM. Inflammatory lung injury is associated with endothelial cell mitochondrial fission and requires the nitration of RhoA and cytoskeletal remodeling. Free Radic Biol Med 2024; 221:125-135. [PMID: 38734269 PMCID: PMC11179967 DOI: 10.1016/j.freeradbiomed.2024.05.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 04/12/2024] [Accepted: 05/09/2024] [Indexed: 05/13/2024]
Abstract
Higher levels of extracellular nicotinamide phosphoribosyltransferase (eNAMPT), a TLR4 agonist, are associated with poor clinical outcomes in sepsis-induced acute lung injury (ALI). Little is known regarding the mechanisms by which eNAMPT is involved in ALI. Our recent work has identified a crucial role for mitochondrial dysfunction in ALI. Thus, this study aimed to determine if eNAMPT-mediated inflammatory injury is associated with the loss of mitochondrial function. Our data show that eNAMPT disrupted mitochondrial bioenergetics. This was associated with cytoskeleton remodeling and the loss of endothelial barrier integrity. These changes were associated with enhanced mitochondrial fission and blocked when Rho-kinase (ROCK) was inhibited. The increases in mitochondrial fission were also associated with the nitration-mediated activation of the small GTPase activator of ROCK, RhoA. Blocking RhoA nitration decreased eNAMPT-mediated mitochondrial fission and endothelial barrier dysfunction. The increase in fission was linked to a RhoA-ROCK mediated increase in Drp1 (dynamin-related protein 1) at serine(S)616. Another TLR4 agonist, lipopolysaccharide (LPS), also increased mitochondrial fission in a Drp1 and RhoA-ROCK-dependent manner. To validate our findings in vivo, we challenged C57BL/6 mice with eNAMPT in the presence and absence of the Drp1 inhibitor, Mdivi-1. Mdivi-1 treatment protected against eNAMPT-induced lung inflammation, edema, and lung injury. These studies demonstrate that mitochondrial fission-dependent disruption of mitochondrial function is essential in TLR4-mediated inflammatory lung injury and identify a key role for RhoA-ROCK signaling. Reducing mitochondrial fission could be a potential therapeutic strategy to improve ARDS outcomes.
Collapse
Affiliation(s)
- Marissa D Pokharel
- Department of Cellular & Molecular Medicine, Herbert Wertheim College of Medicine, Miami, FL, USA; Center for Translational Science, Florida International University, Port St. Lucie, FL, USA
| | - Panfeng Fu
- Center for Translational Science, Florida International University, Port St. Lucie, FL, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, University Park, FL, USA
| | | | - Manivannan Yegambaram
- Center for Translational Science, Florida International University, Port St. Lucie, FL, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, University Park, FL, USA
| | - Qing Lu
- Center for Translational Science, Florida International University, Port St. Lucie, FL, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, University Park, FL, USA
| | - Xutong Sun
- Center for Translational Science, Florida International University, Port St. Lucie, FL, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, University Park, FL, USA
| | - Hoshang Unwalla
- Department of Immunology and Nano-Medicine, Howard Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA
| | - Saurabh Aggarwal
- Department of Cellular & Molecular Medicine, Herbert Wertheim College of Medicine, Miami, FL, USA
| | - Jeffrey R Fineman
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, 94143, USA; Department of Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Ting Wang
- Center for Translational Science, Florida International University, Port St. Lucie, FL, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, University Park, FL, USA
| | - Stephen M Black
- Department of Cellular & Molecular Medicine, Herbert Wertheim College of Medicine, Miami, FL, USA; Center for Translational Science, Florida International University, Port St. Lucie, FL, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, University Park, FL, USA.
| |
Collapse
|
4
|
Balraj P, Ambhore NS, Ramakrishnan YS, Borkar NA, Banerjee P, Reza MI, Varadharajan S, Kumar A, Pabelick CM, Prakash YS, Sathish V. Kisspeptin/KISS1R Signaling Modulates Human Airway Smooth Muscle Cell Migration. Am J Respir Cell Mol Biol 2024; 70:507-518. [PMID: 38512807 PMCID: PMC11160419 DOI: 10.1165/rcmb.2023-0469oc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 03/20/2024] [Indexed: 03/23/2024] Open
Abstract
Airway remodeling is a cardinal feature of asthma, associated with increased airway smooth muscle (ASM) cell mass and upregulation of extracellular matrix deposition. Exaggerated ASM cell migration contributes to excessive ASM mass. Previously, we demonstrated the alleviating role of Kp (kisspeptin) receptor (KISS1R) activation by Kp-10 in mitogen (PDGF [platelet-derived growth factor])-induced human ASM cell proliferation in vitro and airway remodeling in vivo in a mouse model of asthma. Here, we examined the mechanisms by which KISS1R activation regulates mitogen-induced ASM cell migration. KISS1R activation using Kp-10 significantly inhibited PDGF-induced ASM cell migration, further confirmed using KISS1R shRNA. Furthermore, KISS1R activation modulated F/G actin dynamics and the expression of promigration proteins like CDC42 (cell division control protein 42) and cofilin. Mechanistically, we observed reduced ASM RhoA-GTPAse with KISS1R activation. The antimigratory effect of KISS1R was abolished by PKA (protein kinase A)-inhibitory peptide. Conversely, KISS1R activation significantly increased cAMP and phosphorylation of CREB (cAMP-response element binding protein) in PDGF-exposed ASM cells. Overall, these results highlight the alleviating properties of Kp-10 in the context of airway remodeling.
Collapse
Affiliation(s)
- Premanand Balraj
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota
| | | | | | | | - Priyanka Banerjee
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota
| | - Mohammad Irshad Reza
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota
| | - Subashini Varadharajan
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota
| | - Ashish Kumar
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota
| | - Christina M. Pabelick
- Department of Anesthesiology and Perioperative Medicine, and
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Y. S. Prakash
- Department of Anesthesiology and Perioperative Medicine, and
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Venkatachalem Sathish
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota
| |
Collapse
|
5
|
Huang Q, Le Y, Li S, Bian Y. Signaling pathways and potential therapeutic targets in acute respiratory distress syndrome (ARDS). Respir Res 2024; 25:30. [PMID: 38218783 PMCID: PMC10788036 DOI: 10.1186/s12931-024-02678-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 01/03/2024] [Indexed: 01/15/2024] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a common condition associated with critically ill patients, characterized by bilateral chest radiographical opacities with refractory hypoxemia due to noncardiogenic pulmonary edema. Despite significant advances, the mortality of ARDS remains unacceptably high, and there are still no effective targeted pharmacotherapeutic agents. With the outbreak of coronavirus disease 19 worldwide, the mortality of ARDS has increased correspondingly. Comprehending the pathophysiology and the underlying molecular mechanisms of ARDS may thus be essential to developing effective therapeutic strategies and reducing mortality. To facilitate further understanding of its pathogenesis and exploring novel therapeutics, this review provides comprehensive information of ARDS from pathophysiology to molecular mechanisms and presents targeted therapeutics. We first describe the pathogenesis and pathophysiology of ARDS that involve dysregulated inflammation, alveolar-capillary barrier dysfunction, impaired alveolar fluid clearance and oxidative stress. Next, we summarize the molecular mechanisms and signaling pathways related to the above four aspects of ARDS pathophysiology, along with the latest research progress. Finally, we discuss the emerging therapeutic strategies that show exciting promise in ARDS, including several pharmacologic therapies, microRNA-based therapies and mesenchymal stromal cell therapies, highlighting the pathophysiological basis and the influences on signal transduction pathways for their use.
Collapse
Affiliation(s)
- Qianrui Huang
- Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095, Jie Fang Avenue, Wuhan, 430030, China
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jie Fang Avenue, Wuhan, 430030, China
| | - Yue Le
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, 87 Dingjia Bridge, Hunan Road, Gu Lou District, Nanjing, 210009, China
| | - Shusheng Li
- Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095, Jie Fang Avenue, Wuhan, 430030, China.
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jie Fang Avenue, Wuhan, 430030, China.
| | - Yi Bian
- Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095, Jie Fang Avenue, Wuhan, 430030, China.
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jie Fang Avenue, Wuhan, 430030, China.
| |
Collapse
|
6
|
Vielmuth F, Radeva MY, Yeruva S, Sigmund AM, Waschke J. cAMP: A master regulator of cadherin-mediated binding in endothelium, epithelium and myocardium. Acta Physiol (Oxf) 2023; 238:e14006. [PMID: 37243909 DOI: 10.1111/apha.14006] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 05/05/2023] [Accepted: 05/22/2023] [Indexed: 05/29/2023]
Abstract
Regulation of cadherin-mediated cell adhesion is crucial not only for maintaining tissue integrity and barrier function in the endothelium and epithelium but also for electromechanical coupling within the myocardium. Therefore, loss of cadherin-mediated adhesion causes various disorders, including vascular inflammation and desmosome-related diseases such as the autoimmune blistering skin dermatosis pemphigus and arrhythmogenic cardiomyopathy. Mechanisms regulating cadherin-mediated binding contribute to the pathogenesis of diseases and may also be used as therapeutic targets. Over the last 30 years, cyclic adenosine 3',5'-monophosphate (cAMP) has emerged as one of the master regulators of cell adhesion in endothelium and, more recently, also in epithelial cells as well as in cardiomyocytes. A broad spectrum of experimental models from vascular physiology and cell biology applied by different generations of researchers provided evidence that not only cadherins of endothelial adherens junctions (AJ) but also desmosomal contacts in keratinocytes and the cardiomyocyte intercalated discs are central targets in this scenario. The molecular mechanisms involve protein kinase A- and exchange protein directly activated by cAMP-mediated regulation of Rho family GTPases and S665 phosphorylation of the AJ and desmosome adaptor protein plakoglobin. In line with this, phosphodiesterase 4 inhibitors such as apremilast have been proposed as a therapeutic strategy to stabilize cadherin-mediated adhesion in pemphigus and may also be effective to treat other disorders where cadherin-mediated binding is compromised.
Collapse
Affiliation(s)
- Franziska Vielmuth
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Mariya Y Radeva
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Sunil Yeruva
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Anna M Sigmund
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Jens Waschke
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, LMU Munich, Munich, Germany
| |
Collapse
|
7
|
Zemskov EA, Wu X, Aggarwal S, Yegambaram M, Gross C, Lu Q, Wang H, Tang H, Wang T, Black SM. Nitration of protein kinase G-Iα modulates cyclic nucleotide crosstalk via phosphodiesterase 3A: Implications for acute lung injury. J Biol Chem 2021; 297:100946. [PMID: 34252457 PMCID: PMC8342797 DOI: 10.1016/j.jbc.2021.100946] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 06/22/2021] [Accepted: 07/08/2021] [Indexed: 12/05/2022] Open
Abstract
Phosphodiesterase 3A (PDE3A) selectively cleaves the phosphodiester bond of cAMP and is inhibited by cGMP, making it an important regulator of cAMP-cGMP signaling crosstalk in the pulmonary vasculature. In addition, the nitric oxide-cGMP axis is known to play an important role in maintaining endothelial barrier function. However, the potential role of protein kinase G-Iα (PKG-Iα) in this protective process is unresolved and was the focus of our study. We describe here a novel mechanism regulating PDE3A activity, which involves a PKG-Iα-dependent inhibitory phosphorylation of PDE3A at serine 654. We also show that this phosphorylation is critical for maintaining intracellular cAMP levels in the pulmonary endothelium and endothelial barrier integrity. In an animal model of acute lung injury (ALI) induced by challenging mice with lipopolysaccharide (LPS), an increase in PDE3 activity and a decrease in cAMP levels in lung tissue was associated with reduced PKG activity upon PKG-Iα nitration at tyrosine 247. The peroxynitrite scavenger manganese (III) tetrakis(1-methyl-4-pyridyl)porphyrin prevented this increase in PDE3 activity in LPS-exposed lungs. In addition, site-directed mutagenesis of PDE3A to replace serine 654 with alanine yielded a mutant protein that was insensitive to PKG-dependent regulation. Taken together, our data demonstrate a novel functional link between nitrosative stress induced by LPS during ALI and the downregulation of barrier-protective intracellular cAMP levels. Our data also provide new evidence that PKG-Iα is critical for endothelial barrier maintenance and that preservation of its catalytic activity may be efficacious in ALI therapy.
Collapse
Affiliation(s)
- Evgeny A Zemskov
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona, USA
| | - Xiaomin Wu
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona, USA
| | - Saurabh Aggarwal
- Vascular Biology Center, Augusta University, Augusta, Georgia, USA
| | | | - Christine Gross
- Vascular Biology Center, Augusta University, Augusta, Georgia, USA
| | - Qing Lu
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona, USA
| | - Hui Wang
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona, USA; College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Haiyang Tang
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona, USA; College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China; Center for Translational Science, Florida International University, Port Saint Lucie, Florida, USA
| | - Ting Wang
- Center for Translational Science, Florida International University, Port Saint Lucie, Florida, USA; Department of Internal Medicine, The University of Arizona, Phoenix, Arizona, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, Florida, USA
| | - Stephen M Black
- Center for Translational Science, Florida International University, Port Saint Lucie, Florida, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, Florida, USA; Cellular Biology & Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida, USA.
| |
Collapse
|
8
|
Purinergic Regulation of Endothelial Barrier Function. Int J Mol Sci 2021; 22:ijms22031207. [PMID: 33530557 PMCID: PMC7865261 DOI: 10.3390/ijms22031207] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/10/2021] [Accepted: 01/22/2021] [Indexed: 12/12/2022] Open
Abstract
Increased vascular permeability is a hallmark of several cardiovascular anomalies, including ischaemia/reperfusion injury and inflammation. During both ischaemia/reperfusion and inflammation, massive amounts of various nucleotides, particularly adenosine 5'-triphosphate (ATP) and adenosine, are released that can induce a plethora of signalling pathways via activation of several purinergic receptors and may affect endothelial barrier properties. The nature of the effects on endothelial barrier function may depend on the prevalence and type of purinergic receptors activated in a particular tissue. In this review, we discuss the influence of the activation of various purinergic receptors and downstream signalling pathways on vascular permeability during pathological conditions.
Collapse
|
9
|
RAC1 nitration at Y 32 IS involved in the endothelial barrier disruption associated with lipopolysaccharide-mediated acute lung injury. Redox Biol 2020; 38:101794. [PMID: 33248422 PMCID: PMC7664366 DOI: 10.1016/j.redox.2020.101794] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 11/06/2020] [Accepted: 11/07/2020] [Indexed: 02/06/2023] Open
Abstract
Acute lung injury (ALI), a devastating illness induced by systemic inflammation e.g., sepsis or local lung inflammation e.g., COVID-19 mediated severe pneumonia, has an unacceptably high mortality and has no effective therapy. ALI is associated with increased pulmonary microvascular hyperpermeability and alveolar flooding. The small Rho GTPases, RhoA and Rac1 are central regulators of vascular permeability through cytoskeleton rearrangements. RhoA and Rac1 have opposing functional outcome: RhoA induces an endothelial contractile phenotype and barrier disruption, while Rac1 stabilizes endothelial junctions and increases barrier integrity. In ALI, RhoA activity is increased while Rac1 activity is reduced. We have shown that the activation of RhoA in lipopolysaccharide (LPS)-mediated ALI, is dependent, at least in part, on a single nitration event at tyrosine (Y)34. Thus, the purpose of this study was to determine if the inhibition of Rac1 is also dependent on its nitration. Our data show that Rac1 inhibition by LPS is associated with its nitration that mass spectrometry identified as Y32, within the switch I region adjacent to the nucleotide-binding site. Using a molecular modeling approach, we designed a nitration shielding peptide for Rac1, designated NipR2 (nitration inhibitor peptide for the Rho GTPases 2), which attenuated the LPS-induced nitration of Rac1 at Y32, preserves Rac1 activity and attenuates the LPS-mediated disruption of the endothelial barrier in human lung microvascular endothelial cells (HLMVEC). Using a murine model of ALI induced by intratracheal installation of LPS we found that NipR2 successfully prevented Rac1 nitration and Rac1 inhibition, and more importantly attenuated pulmonary inflammation, reduced lung injury and prevented the loss of lung function. Together, our data identify a new post-translational mechanism of Rac1 inhibition through its nitration at Y32. As NipR2 also reduces sepsis induced ALI in the mouse lung, we conclude that Rac1 nitration is a therapeutic target in ALI. Endotoxin exposure induces site specific nitration of Rac1 at Y32 via peroxynitrite stress. Rac1 nitration at Y32 leads to persistent Rac GTPase inhibition and endothelial barrier disruption. Novel Rac1 nitration shielding peptide, NipR2 blocks Rac1 nitration and rescues endotoxin induced lung inflammation. NipR2 is potentially an effective therapy for sepsis induced lung injury by targeting Rac1 Y32 nitration.
Collapse
|
10
|
Gündüz D, Troidl C, Tanislav C, Rohrbach S, Hamm C, Aslam M. Role of PI3K/Akt and MEK/ERK Signalling in cAMP/Epac-Mediated Endothelial Barrier Stabilisation. Front Physiol 2019; 10:1387. [PMID: 31787905 PMCID: PMC6855264 DOI: 10.3389/fphys.2019.01387] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Accepted: 10/22/2019] [Indexed: 12/16/2022] Open
Abstract
Background and Aims Activation of the cAMP/Epac signalling stabilises endothelial barrier function. Moreover, its activation is accompanied by an activation of PI3K/Akt and MEK/ERK signalling in diverse cell types but their impact on endothelial barrier function is largely unknown. Here the role of PI3K/Akt and MEK/ERK signalling in cAMP/Epac-mediated endothelial barrier stabilisation was analysed. Methods Endothelial barrier function was analysed in cultured human umbilical vein endothelial cells (HUVECs) by measuring flux of albumin. A modified cAMP analogue 8-pCPT-2′-O-Me-cAMP (Epac agonist) was used to specifically activate cAMP/Epac signalling. Results Epac agonist reduces the basal and attenuates thrombin-induced endothelial hyperpermeability accompanied by an activation of PI3K/Akt and MEK/ERK signalling. The qPCR data demonstrate HUVECs express PI3Kα, PI3Kβ, and PI3Kγ but not PI3Kδ isoforms. The western blot data demonstrate Epac agonist activates PI3Kα and PI3Kβ isoforms. Inhibition of MEK/ERK but not PI3K/Akt pathway potentiates the endothelial barrier protective effects of cAMP/Epac signalling. Inhibition of MEK/ERK signalling in the presence of Epac agonist induces a reorganisation of actin cytoskeleton to the cell periphery, enhanced VE-cadherin localisation at cell-cell junctions, and dephosphorylation of myosin light chains (MLC) but not inhibition of RhoA/Rock signalling. Moreover, Epac agonist promotes endothelial cell (EC) survival via reduction in activities of pro-apoptotic caspases in a PI3K/Akt and MEK/ERK signalling-dependent manner. Conclusion Our data demonstrate that the Epac agonist simultaneously activates diverse signalling pathways in ECs, which may have differential effects on endothelial barrier function. It activates PI3K/Akt and MEK/ERK signalling which mainly govern its pro-survival effects on ECs. Inhibition of MEK/ERK but not PI3K/Akt signalling enhances barrier stabilising and barrier protective effects of cAMP/Epac activation. Chemical Compounds Used In This Study 8-pCPT-2′-O-Me-cAMP (PubChem CID: 9913268); Akt inhibitor VIII (PubChem CID: 10196499); AS-252424 (PubChem CID: 11630874); IC-87114 (PubChem CID: 9908783); PD 98059 (PubChem CID: 4713); PIK-75 (PubChem CID: 10275789); TGX-221 (PubChem CID: 9907093); Thrombin (PubChem CID: 90470996); U0126 (PubChem CID: 3006531); Wortmannin (PubChem CID: 312145).
Collapse
Affiliation(s)
- Dursun Gündüz
- Department of Cardiology and Angiology, University Hospital of Giessen and Marburg, Giessen, Germany.,Department of Cardiology and Angiology, Evangelisches Jung Stilling Krankenhaus GmbH, Siegen, Germany
| | - Christian Troidl
- Department of Cardiology and Angiology, University Hospital of Giessen and Marburg, Giessen, Germany.,Experimental Cardiology, Justus Liebig University Giessen, Giessen, Germany
| | - Christian Tanislav
- Department of Neurology, Evangelisches Jung Stilling Krankenhaus GmbH, Siegen, Germany.,Department of Neurology, University Hospital of Giessen and Marburg, Giessen, Germany
| | - Susanne Rohrbach
- Institute of Physiology, Justus Liebig University Giessen, Giessen, Germany
| | - Christian Hamm
- Department of Cardiology and Angiology, University Hospital of Giessen and Marburg, Giessen, Germany
| | - Muhammad Aslam
- Department of Cardiology and Angiology, University Hospital of Giessen and Marburg, Giessen, Germany.,Experimental Cardiology, Justus Liebig University Giessen, Giessen, Germany
| |
Collapse
|
11
|
Sun ZL, Jiang XF, Cheng YC, Liu YF, Yang K, Zhu SL, Kong XB, Tu Y, Bian KF, Liu ZL, Chen XY. Exendin-4 inhibits high-altitude cerebral edema by protecting against neurobiological dysfunction. Neural Regen Res 2018; 13:653-663. [PMID: 29722317 PMCID: PMC5950675 DOI: 10.4103/1673-5374.230291] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The anti-inflammatory and antioxidant effects of exendin-4 (Ex-4) have been reported previously. However, whether (Ex-4) has anti-inflammatory and antioxidant effects on high-altitude cerebral edema (HACE) remains poorly understood. In this study, two rat models of HACE were established by placing rats in a hypoxic environment with a simulated altitude of either 6000- or 7000-m above sea level (MASL) for 72 hours. An altitude of 7000 MASL with 72-hours of hypoxia was found to be the optimized experimental paradigm for establishing HACE models. Then, in rats where a model of HACE was established by introducing them to a 7000 MASL environment with 72-hours of hypoxia treatment, 2, 10 and, 100 μg of Ex-4 was intraperitoneally administrated. The open field test and tail suspension test were used to test animal behavior. Routine methods were used to detect change in inflammatory cells. Hematoxylin-eosin staining was performed to determine pathological changes to brain tissue. Wet/dry weight ratios were used to measure brain water content. Evans blue leakage was used to determine blood-brain barrier integrity. Enzyme-linked immunosorbent assay (ELISA) was performed to measure markers of inflammation and oxidative stress including superoxide dismutase, glutathione, and malonaldehyde values, as well as interleukin-6, tumor necrosis factor-alpha, cyclic adenosine monophosphate levels in the brain tissue. Western blot analysis was performed to determine the levels of occludin, ZO-1, SOCS-3, vascular endothelial growth factor, EPAC1, nuclear factor-kappa B, and aquaporin-4. Our results demonstrate that Ex-4 preconditioning decreased brain water content, inhibited inflammation and oxidative stress, alleviated brain tissue injury, maintain blood-brain barrier integrity, and effectively improved motor function in rat models of HACE. These findings suggest that Ex-4 exhibits therapeutic potential in the treatment of HACE.
Collapse
Affiliation(s)
- Zhong-Lei Sun
- Affiliated Hospital of Logistics University of Chinese People's Armed Police Forces, Institute of Traumatic Brain Trauma and Neurological of CAPF, Neurotrauma Repair Key Laboratory of Tianjin, Tianjin; Jinzhou Medical University, Jinzhou, Liaoning Province, China
| | - Xian-Feng Jiang
- Affiliated Hospital of Logistics University of Chinese People's Armed Police Forces, Institute of Traumatic Brain Trauma and Neurological of CAPF, Neurotrauma Repair Key Laboratory of Tianjin; Tianjin Medical University, Tianjin, China
| | - Yuan-Chi Cheng
- Central Hospital of Fengxian District of Shanghai, Shanghai, China
| | - Ying-Fu Liu
- Logistics University of People's Armed Police Force, Tianjin, China
| | - Kai Yang
- The No. 2 Hospital of Nanjing, Nanjing, Jiangsu Province, China
| | | | - Xian-Bin Kong
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yue Tu
- Affiliated Hospital of Logistics University of Chinese People's Armed Police Forces, Institute of Traumatic Brain Trauma and Neurological of CAPF, Neurotrauma Repair Key Laboratory of Tianjin, Tianjin, China
| | - Ke-Feng Bian
- Logistics University of People's Armed Police Force, Tianjin, China
| | - Zhen-Lin Liu
- Affiliated Hospital of Logistics University of Chinese People's Armed Police Forces, Institute of Traumatic Brain Trauma and Neurological of CAPF, Neurotrauma Repair Key Laboratory of Tianjin, Tianjin, China
| | - Xu-Yi Chen
- Affiliated Hospital of Logistics University of Chinese People's Armed Police Forces, Institute of Traumatic Brain Trauma and Neurological of CAPF, Neurotrauma Repair Key Laboratory of Tianjin, Tianjin, China
| |
Collapse
|
12
|
Breitbart H, Finkelstein M. Actin cytoskeleton and sperm function. Biochem Biophys Res Commun 2017; 506:372-377. [PMID: 29102633 DOI: 10.1016/j.bbrc.2017.11.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 11/01/2017] [Indexed: 11/17/2022]
Abstract
For the acquisition of the ability to fertilize the egg, mammalian spermatozoa should undergo a series of biochemical transformations in the female reproductive tract, collectively called capacitation. The capacitated sperm can undergo the acrosomal exocytosis process near or on the oocyte, which allows the spermatozoon to penetrate and fertilize it. One of the main processes in capacitation involves dynamic cytoskeletal remodeling particularly of actin. Actin polymerization occurs during sperm capacitation and the produced F-actin should be depolymerized prior to the acrosomal exocytosis. In the present review, we describe the mechanisms that regulate F-actin formation during sperm capacitation and the F-actin dispersion prior to the acrosomal exocytosis. During sperm capacitation, the actin severing proteins gelsolin and cofilin are inactive and they undergo activation prior to the acrosomal exocytosis.
Collapse
Affiliation(s)
- Haim Breitbart
- The Mina & Everard Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 5290002, Israel.
| | | |
Collapse
|
13
|
Milberg O, Shitara A, Ebrahim S, Masedunskas A, Tora M, Tran DT, Chen Y, Conti MA, Adelstein RS, Ten Hagen KG, Weigert R. Concerted actions of distinct nonmuscle myosin II isoforms drive intracellular membrane remodeling in live animals. J Cell Biol 2017; 216:1925-1936. [PMID: 28600434 PMCID: PMC5496622 DOI: 10.1083/jcb.201612126] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 04/02/2017] [Accepted: 05/02/2017] [Indexed: 12/11/2022] Open
Abstract
Membrane remodeling plays a fundamental role during a variety of biological events. However, the dynamics and the molecular mechanisms regulating this process within cells in mammalian tissues in situ remain largely unknown. In this study, we use intravital subcellular microscopy in live mice to study the role of the actomyosin cytoskeleton in driving the remodeling of membranes of large secretory granules, which are integrated into the plasma membrane during regulated exocytosis. We show that two isoforms of nonmuscle myosin II, NMIIA and NMIIB, control distinct steps of the integration process. Furthermore, we find that F-actin is not essential for the recruitment of NMII to the secretory granules but plays a key role in the assembly and activation of NMII into contractile filaments. Our data support a dual role for the actomyosin cytoskeleton in providing the mechanical forces required to remodel the lipid bilayer and serving as a scaffold to recruit key regulatory molecules.
Collapse
Affiliation(s)
- Oleg Milberg
- Intracellular Membrane Trafficking Section, National Institutes of Health, Bethesda, MD
| | - Akiko Shitara
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD.,Intracellular Membrane Trafficking Section, National Institutes of Health, Bethesda, MD
| | - Seham Ebrahim
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Andrius Masedunskas
- Intracellular Membrane Trafficking Section, National Institutes of Health, Bethesda, MD.,School of Medical Sciences, University of New South Wales, Sidney, Australia
| | - Muhibullah Tora
- Intracellular Membrane Trafficking Section, National Institutes of Health, Bethesda, MD
| | - Duy T Tran
- Developmental Glycobiology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD
| | - Yun Chen
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Mary Anne Conti
- Laboratory of Molecular Cardiology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Robert S Adelstein
- Laboratory of Molecular Cardiology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Kelly G Ten Hagen
- Developmental Glycobiology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD
| | - Roberto Weigert
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD .,Intracellular Membrane Trafficking Section, National Institutes of Health, Bethesda, MD
| |
Collapse
|
14
|
RhoA S-nitrosylation as a regulatory mechanism influencing endothelial barrier function in response to G +-bacterial toxins. Biochem Pharmacol 2016; 127:34-45. [PMID: 28017778 DOI: 10.1016/j.bcp.2016.12.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 12/19/2016] [Indexed: 01/05/2023]
Abstract
Disruption of the endothelial barrier in response to Gram positive (G+) bacterial toxins is a major complication of acute lung injury (ALI) and can be further aggravated by antibiotics which stimulate toxin release. The integrity of the pulmonary endothelial barrier is mediated by the balance of disruptive forces such as the small GTPase RhoA, and protective forces including endothelium-derived nitric oxide (NO). How NO protects against the barrier dysfunction is incompletely understood and our goal was to determine whether NO and S-nitrosylation can modulate RhoA activity and whether this mechanism is important for G+ toxin-induced microvascular permeability. We found that the G+ toxin listeriolysin-O (LLO) increased RhoA activity and that NO and S-NO donors inhibit RhoA activity. RhoA was robustly S-nitrosylated as determined by biotin-switch and mercury column analysis. MS revealed that three primary cysteine residues are S-nitrosylated including cys16, cys20 and cys159. Mutation of these residues to serine diminished S-nitrosylation to endogenous NO and mutant RhoA was less sensitive to inhibition by S-NO. G+-toxins stimulated the denitrosylation of RhoA which was not mediated by S-nitrosoglutathione reductase (GSNOR), thioredoxin (TRX) or thiol-dependent enzyme activity but was instead stimulated directly by elevated calcium levels. Calcium-promoted the direct denitrosylation of WT but not mutant RhoA and mutant RhoA adenovirus was more effective than WT in disrupting the barrier integrity of human lung microvascular endothelial cells. In conclusion, we reveal a novel mechanism by which NO and S-nitrosylation reduces RhoA activity which may be of significance in the management of pulmonary endothelial permeability induced by G+-toxins.
Collapse
|
15
|
Zhao Y, Zhang X, Li J, Bian Y, Sheng M, Liu B, Fu Z, Zhang Y, Yang B. Jujuboside B Reduces Vascular Tension by Increasing Ca2+ Influx and Activating Endothelial Nitric Oxide Synthase. PLoS One 2016; 11:e0149386. [PMID: 26901291 PMCID: PMC4762982 DOI: 10.1371/journal.pone.0149386] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 02/01/2016] [Indexed: 11/24/2022] Open
Abstract
Jujuboside B has been reported to have protective effect on many cardiovascular diseases. However, the effects of Jujuboside B on vascular tension and endothelial function are unknown. The present study investigated the effects of Jujuboside B on reducing vascular tension, protecting endothelial function and the potential mechanisms. The tension of isolated rat thoracic aorta ring was measured by Wire myograph system. The concentration of nitric oxide (NO) and the activity of endothelial nitric oxide synthase (eNOS) in human aortic endothelial cells (HAECs) were determined by Griess reagent method and enzyme-linked immune sorbent assay. The protein levels of eNOS and p-eNOS at Serine-1177 were determined by western blot analysis. Intracellular Ca2+ concentration in HAECs was measured by laser confocal imaging microscopy. Results showed that Jujuboside B reduced the tension of rat thoracic aorta rings with intact endothelium in a dose-dependent manner. L-NAME, KN93, EGTA, SKF96365, iberiotoxin and glibenclamide significantly attenuated Jujuboside B-induced vasodilation in endothelium-intact tissues. In contrast, indometacin and 4-DAMP had no such effects. Jujuboside B also promoted NO generation and increased eNOS activity, which were attenuated by L-NAME, EGTA and SKF96365. Moreover, Jujuboside B increased intracellular Ca2+ concentration dose-dependently, which was inhibited by EGTA and SKF96365. Besides, Jujuboside B induced a rapid Ca2+ influx instantaneously after depleting intracellular Ca2+ store, which was significantly inhibited by SKF96365. In conclusion, this study preliminarily confirmed that Jujuboside B reduced vascular tension endothelium-dependently. The underlying mechanisms involved that Jujuboside B increased extracellular Ca2+ influx through endothelial transient receptor potential cation (TRPC) channels, phosphorylated eNOS and promoted NO generation in vascular endothelial cells. In addition, Jujuboside B-induced vasodilation involved endothelium-dependent hyperpolarizaiton through endothelial potassium channels. Jujuboside B is a natural compound with new pharmacological effects on improving endothelial dysfunction and treating vascular diseases.
Collapse
Affiliation(s)
- Yixiu Zhao
- Department of Pharmacology, Harbin Medical University, Harbin, Heilongjiang, PR China
- Key Laboratory of Cardiovascular Medicine Research (Harbin Medical University), Ministry of Education, Harbin, Heilongjiang, PR China
| | - Xin Zhang
- Department of Pharmacology, Harbin Medical University, Harbin, Heilongjiang, PR China
- Key Laboratory of Cardiovascular Medicine Research (Harbin Medical University), Ministry of Education, Harbin, Heilongjiang, PR China
| | - Jiannan Li
- Department of Pharmacology, Harbin Medical University, Harbin, Heilongjiang, PR China
- Key Laboratory of Cardiovascular Medicine Research (Harbin Medical University), Ministry of Education, Harbin, Heilongjiang, PR China
| | - Yu Bian
- Department of Pharmacology, Harbin Medical University, Harbin, Heilongjiang, PR China
- Key Laboratory of Cardiovascular Medicine Research (Harbin Medical University), Ministry of Education, Harbin, Heilongjiang, PR China
| | - Miaomiao Sheng
- Department of Pharmacology, Harbin Medical University, Harbin, Heilongjiang, PR China
- Key Laboratory of Cardiovascular Medicine Research (Harbin Medical University), Ministry of Education, Harbin, Heilongjiang, PR China
| | - Bin Liu
- Department of Pharmacology, Harbin Medical University, Harbin, Heilongjiang, PR China
- Key Laboratory of Cardiovascular Medicine Research (Harbin Medical University), Ministry of Education, Harbin, Heilongjiang, PR China
| | - Zidong Fu
- Department of Pharmacology, Harbin Medical University, Harbin, Heilongjiang, PR China
- Key Laboratory of Cardiovascular Medicine Research (Harbin Medical University), Ministry of Education, Harbin, Heilongjiang, PR China
| | - Yan Zhang
- Department of Pharmacology, Harbin Medical University, Harbin, Heilongjiang, PR China
- Key Laboratory of Cardiovascular Medicine Research (Harbin Medical University), Ministry of Education, Harbin, Heilongjiang, PR China
- * E-mail: (YZ); (BFY)
| | - Baofeng Yang
- Department of Pharmacology, Harbin Medical University, Harbin, Heilongjiang, PR China
- Key Laboratory of Cardiovascular Medicine Research (Harbin Medical University), Ministry of Education, Harbin, Heilongjiang, PR China
- * E-mail: (YZ); (BFY)
| |
Collapse
|
16
|
Gerarduzzi C, He Q, Antoniou J, Di Battista JA. Prostaglandin E(2)-dependent blockade of actomyosin and stress fibre formation is mediated through S1379 phosphorylation of ROCK2. J Cell Biochem 2015; 115:1516-27. [PMID: 24610576 DOI: 10.1002/jcb.24806] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2013] [Accepted: 03/04/2014] [Indexed: 12/26/2022]
Abstract
Prostaglandin E2 is a pleiotropic bioactive lipid that controls cytoskeletal alterations, although the precise G-protein coupled EP receptor signalling mechanisms remain ill defined. We adopted a phosphoproteomic approach to characterize post-receptor downstream signalling substrates using antibodies that selectively recognize and immunoprecipitate phosphorylated substrates of a number of kinases. Using human synovial fibroblasts in monolayer cell culture, PGE2 induced rapid and sustained changes in cellular morphology and reduction in cytoplasmic volume that were associated with disassembly of the phalloidin-stained stress fibres as judged by light and confocal microscopy. Furthermore, PGE2 induced a rapid dephosphorylation of myosin light chain II (MLC) at S19 under basal or cytokine-induced conditions that was linked to an activation of myosin light chain phosphatase. The use of specific synthetic EP agonists suggested that the response was mediated by EP2 receptors, as other EP agonists did not manifest the same effect on MLC phosphorylation. In addition, PGE2 induced sustained Y118 dephosphorylation of phospho-paxillin and loss of focal adhesions as observed by confocal microscopy and Western analysis. Phosphoproteomic analysis of PGE2 /GPCR/PKA phosphosubstrates identified a unique, non-redundant, phosphorylated (>30-fold) site on rho-associated coiled coil-containing kinase 2 (ROCK2) at S1379. Analysis of ROCK2 mutant behaviour (e.g. S1379A) in overexpression studies revealed that PGE2 -dependent phosphorylation of ROCK2 resulted in the inhibition of the kinase, since induced MLC phosphorylation was no longer blocked by PGE2 nor could PGE2 induce disassembly of stress fibres. Thus, PGE2 -dependent blockade of actomyosin fibre formation, characteristic of myofibroblasts, may be mediated through specific ROCK2 S1379 phosphorylation.
Collapse
Affiliation(s)
- Casimiro Gerarduzzi
- Department of Genetics and Complex Diseases, Harvard School of Public Health, Boston, Massachusetts; Department of Experimental Medicine, McGill University, Montreal, Quebec, Canada
| | | | | | | |
Collapse
|
17
|
Lezhnina K, Kovalchuk O, Zhavoronkov AA, Korzinkin MB, Zabolotneva AA, Shegay PV, Sokov DG, Gaifullin NM, Rusakov IG, Aliper AM, Roumiantsev SA, Alekseev BY, Borisov NM, Buzdin AA. Novel robust biomarkers for human bladder cancer based on activation of intracellular signaling pathways. Oncotarget 2015; 5:9022-32. [PMID: 25296972 PMCID: PMC4253415 DOI: 10.18632/oncotarget.2493] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
We recently proposed a new bioinformatic algorithm called OncoFinder for quantifying the activation of intracellular signaling pathways. It was proved advantageous for minimizing errors of high-throughput gene expression analyses and showed strong potential for identifying new biomarkers. Here, for the first time, we applied OncoFinder for normal and cancerous tissues of the human bladder to identify biomarkers of bladder cancer. Using Illumina HT12v4 microarrays, we profiled gene expression in 17 cancer and seven non-cancerous bladder tissue samples. These experiments were done in two independent laboratories located in Russia and Canada. We calculated pathway activation strength values for the investigated transcriptomes and identified signaling pathways that were regulated differently in bladder cancer (BC) tissues compared with normal controls. We found, for both experimental datasets, 44 signaling pathways that serve as excellent new biomarkers of BC, supported by high area under the curve (AUC) values. We conclude that the OncoFinder approach is highly efficient in finding new biomarkers for cancer. These markers are mathematical functions involving multiple gene products, which distinguishes them from “traditional” expression biomarkers that only assess concentrations of single genes.
Collapse
Affiliation(s)
- Ksenia Lezhnina
- Pathway Pharmaceuticals, Wan Chai, Hong Kong, Hong Kong SAR. Laboratory of Bioinformatics, D. Rogachyov Federal Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Olga Kovalchuk
- Department of Biological Sciences, University of Lethbridge, 4401 University Drive, Lethbridge, AB, T1K 3M4. Canada Cancer and Aging Research Laboratories, Lethbridge, AB, Canada
| | - Alexander A Zhavoronkov
- Laboratory of Bioinformatics, D. Rogachyov Federal Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia. Insilico Medicine, Inc, ETC, Johns Hopkins University, Baltimore, MD. Faculty of Biological and Medical Physics, Moscow Institute of Physics and Technology
| | | | - Anastasia A Zabolotneva
- Group for Genomic Regulation of Cell Signaling Systems, Shemyakn-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
| | - Peter V Shegay
- P.A. Herzen Moscow Oncological Research Institute, Moscow, Russia
| | | | - Nurshat M Gaifullin
- Lomonosov Moscow State University, Faculty of Fundamental Medicine, Moscow, Russia. Russian medical postgraduate academy,Moscow, Russia
| | - Igor G Rusakov
- P.A. Herzen Moscow Oncological Research Institute, Moscow, Russia
| | - Alexander M Aliper
- Pathway Pharmaceuticals, Wan Chai, Hong Kong, Hong Kong SAR. Laboratory of Bioinformatics, D. Rogachyov Federal Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Sergey A Roumiantsev
- Laboratory of Bioinformatics, D. Rogachyov Federal Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Boris Y Alekseev
- P.A. Herzen Moscow Oncological Research Institute, Moscow, Russia
| | - Nikolay M Borisov
- Laboratory of Systems Biology, A.I. Burnasyan Federal Medical Biophysical Center, Moscow, Russia
| | - Anton A Buzdin
- Pathway Pharmaceuticals, Wan Chai, Hong Kong, Hong Kong SAR. Laboratory of Bioinformatics, D. Rogachyov Federal Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia. Group for Genomic Regulation of Cell Signaling Systems, Shemyakn-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
| |
Collapse
|
18
|
The role and importance of cofilin in human sperm capacitation and the acrosome reaction. Cell Tissue Res 2015; 362:665-75. [DOI: 10.1007/s00441-015-2229-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 05/20/2015] [Indexed: 10/23/2022]
|
19
|
Arbeille E, Reynaud F, Sanyas I, Bozon M, Kindbeiter K, Causeret F, Pierani A, Falk J, Moret F, Castellani V. Cerebrospinal fluid-derived Semaphorin3B orients neuroepithelial cell divisions in the apicobasal axis. Nat Commun 2015; 6:6366. [PMID: 25721514 DOI: 10.1038/ncomms7366] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 01/22/2015] [Indexed: 01/05/2023] Open
Abstract
The spatial orientation of cell divisions is fundamental for tissue architecture and homeostasis. Here we analysed neuroepithelial progenitors in the developing mouse spinal cord to determine whether extracellular signals orient the mitotic spindle. We report that Semaphorin3B (Sema3B) released from the floor plate and the nascent choroid plexus in the cerebrospinal fluid (CSF) controls progenitor division orientation. Delivery of exogenous Sema3B to neural progenitors after neural tube opening in living embryos promotes planar orientation of their division. Preventing progenitor access to cues present in the CSF by genetically engineered canal obstruction affects the proportion of planar and oblique divisions. Sema3B knockout phenocopies the loss of progenitor access to the CSF. Sema3B binds to the apical surface of mitotic progenitors and exerts its effect via Neuropilin receptors, GSK3 activation and subsequent inhibition of the microtubule stabilizer CRMP2. Thus, extrinsic control mediated by the Semaphorin signalling orients progenitor divisions in neurogenic zones.
Collapse
Affiliation(s)
- Elise Arbeille
- University of Lyon, University of Lyon1, CGΦMC, UMR CNRS 5534, F-69100 Villeurbanne, France
| | - Florie Reynaud
- University of Lyon, University of Lyon1, CGΦMC, UMR CNRS 5534, F-69100 Villeurbanne, France
| | - Isabelle Sanyas
- University of Lyon, University of Lyon1, CGΦMC, UMR CNRS 5534, F-69100 Villeurbanne, France
| | - Muriel Bozon
- University of Lyon, University of Lyon1, CGΦMC, UMR CNRS 5534, F-69100 Villeurbanne, France
| | - Karine Kindbeiter
- University of Lyon, University of Lyon1, CGΦMC, UMR CNRS 5534, F-69100 Villeurbanne, France
| | - Frédéric Causeret
- CNRS UMR 7592, Institut Jacques Monod, University Paris Diderot, Sorbonne Paris Cité, F-75205 Paris, France
| | - Alessandra Pierani
- CNRS UMR 7592, Institut Jacques Monod, University Paris Diderot, Sorbonne Paris Cité, F-75205 Paris, France
| | - Julien Falk
- University of Lyon, University of Lyon1, CGΦMC, UMR CNRS 5534, F-69100 Villeurbanne, France
| | - Frédéric Moret
- University of Lyon, University of Lyon1, CGΦMC, UMR CNRS 5534, F-69100 Villeurbanne, France
| | - Valérie Castellani
- University of Lyon, University of Lyon1, CGΦMC, UMR CNRS 5534, F-69100 Villeurbanne, France
| |
Collapse
|
20
|
Pierozan P, Ferreira F, Ortiz de Lima B, Gonçalves Fernandes C, Totarelli Monteforte P, de Castro Medaglia N, Bincoletto C, Soubhi Smaili S, Pessoa-Pureur R. The phosphorylation status and cytoskeletal remodeling of striatal astrocytes treated with quinolinic acid. Exp Cell Res 2014; 322:313-23. [PMID: 24583400 DOI: 10.1016/j.yexcr.2014.02.024] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Revised: 01/10/2014] [Accepted: 02/19/2014] [Indexed: 12/24/2022]
Abstract
Quinolinic acid (QUIN) is a glutamate agonist which markedly enhances the vulnerability of neural cells to excitotoxicity. QUIN is produced from the amino acid tryptophan through the kynurenine pathway (KP). Dysregulation of this pathway is associated with neurodegenerative conditions. In this study we treated striatal astrocytes in culture with QUIN and assayed the endogenous phosphorylating system associated with glial fibrillary acidic protein (GFAP) and vimentin as well as cytoskeletal remodeling. After 24h incubation with 100 µM QUIN, cells were exposed to (32)P-orthophosphate and/or protein kinase A (PKA), protein kinase dependent of Ca(2+)/calmodulin II (PKCaMII) or protein kinase C (PKC) inhibitors, H89 (20 μM), KN93 (10 μM) and staurosporin (10nM), respectively. Results showed that hyperphosphorylation was abrogated by PKA and PKC inhibitors but not by the PKCaMII inhibitor. The specific antagonists to ionotropic NMDA and non-NMDA (50 µM DL-AP5 and CNQX, respectively) glutamate receptors as well as to metabotropic glutamate receptor (mGLUR; 50 µM MCPG), mGLUR1 (100 µM MPEP) and mGLUR5 (10 µM 4C3HPG) prevented the hyperphosphorylation provoked by QUIN. Also, intra and extracellular Ca(2+) quelators (1mM EGTA; 10 µM BAPTA-AM, respectively) prevented QUIN-mediated effect, while Ca(2+) influx through voltage-dependent Ca(2+) channel type L (L-VDCC) (blocker: 10 µM verapamil) is not implicated in this effect. Morphological analysis showed dramatically altered actin cytoskeleton with concomitant change of morphology to fusiform and/or flattened cells with retracted cytoplasm and disruption of the GFAP meshwork, supporting misregulation of actin cytoskeleton. Both hyperphosphorylation and cytoskeletal remodeling were reversed 24h after QUIN removal. Astrocytes are highly plastic cells and the vulnerability of astrocyte cytoskeleton may have important implications for understanding the neurotoxicity of QUIN in neurodegenerative disorders.
Collapse
Affiliation(s)
- Paula Pierozan
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS 90035-003, Brazil
| | - Fernanda Ferreira
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS 90035-003, Brazil
| | - Bárbara Ortiz de Lima
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS 90035-003, Brazil
| | - Carolina Gonçalves Fernandes
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS 90035-003, Brazil
| | | | | | - Claudia Bincoletto
- Departamento de Farmacologia, Universidade Federal de São Paulo (UNIFESP/EPM), São Paulo, SP, Brazil
| | - Soraya Soubhi Smaili
- Departamento de Farmacologia, Universidade Federal de São Paulo (UNIFESP/EPM), São Paulo, SP, Brazil
| | - Regina Pessoa-Pureur
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS 90035-003, Brazil.
| |
Collapse
|
21
|
Rafikov R, Dimitropoulou C, Aggarwal S, Kangath A, Gross C, Pardo D, Sharma S, Jezierska-Drutel A, Patel V, Snead C, Lucas R, Verin A, Fulton D, Catravas JD, Black SM. Lipopolysaccharide-induced lung injury involves the nitration-mediated activation of RhoA. J Biol Chem 2014; 289:4710-22. [PMID: 24398689 DOI: 10.1074/jbc.m114.547596] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Acute lung injury (ALI) is characterized by increased endothelial hyperpermeability. Protein nitration is involved in the endothelial barrier dysfunction in LPS-exposed mice. However, the nitrated proteins involved in this process have not been identified. The activation of the small GTPase RhoA is a critical event in the barrier disruption associated with LPS. Thus, in this study we evaluated the possible role of RhoA nitration in this process. Mass spectroscopy identified a single nitration site, located at Tyr(34) in RhoA. Tyr(34) is located within the switch I region adjacent to the nucleotide-binding site. Utilizing this structure, we developed a peptide designated NipR1 (nitration inhibitory peptide for RhoA 1) to shield Tyr(34) against nitration. TAT-fused NipR1 attenuated RhoA nitration and barrier disruption in LPS-challenged human lung microvascular endothelial cells. Further, treatment of mice with NipR1 attenuated vessel leakage and inflammatory cell infiltration and preserved lung function in a mouse model of ALI. Molecular dynamics simulations suggested that the mechanism by which Tyr(34) nitration stimulates RhoA activity was through a decrease in GDP binding to the protein caused by a conformational change within a region of Switch I, mimicking the conformational shift observed when RhoA is bound to a guanine nucleotide exchange factor. Stopped flow kinetic analysis was used to confirm this prediction. Thus, we have identified a new mechanism of nitration-mediated RhoA activation involved in LPS-mediated endothelial barrier dysfunction and show the potential utility of "shielding" peptides to prevent RhoA nitration in the management of ALI.
Collapse
Affiliation(s)
- Ruslan Rafikov
- From the Program in Pulmonary Vascular Disease, Vascular Biology Center and
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Arnold KM, Goeckeler ZM, Wysolmerski RB. Loss of Focal Adhesion Kinase Enhances Endothelial Barrier Function and Increases Focal Adhesions. Microcirculation 2013; 20:637-49. [DOI: 10.1111/micc.12063] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Accepted: 04/16/2013] [Indexed: 12/30/2022]
|
23
|
Oldenburger A, Maarsingh H, Schmidt M. Multiple facets of cAMP signalling and physiological impact: cAMP compartmentalization in the lung. Pharmaceuticals (Basel) 2012; 5:1291-331. [PMID: 24281338 PMCID: PMC3816672 DOI: 10.3390/ph5121291] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Revised: 11/15/2012] [Accepted: 11/20/2012] [Indexed: 12/20/2022] Open
Abstract
Therapies involving elevation of the endogenous suppressor cyclic AMP (cAMP) are currently used in the treatment of several chronic inflammatory disorders, including chronic obstructive pulmonary disease (COPD). Characteristics of COPD are airway obstruction, airway inflammation and airway remodelling, processes encompassed by increased airway smooth muscle mass, epithelial changes, goblet cell and submucosal gland hyperplasia. In addition to inflammatory cells, airway smooth muscle cells and (myo)fibroblasts, epithelial cells underpin a variety of key responses in the airways such as inflammatory cytokine release, airway remodelling, mucus hypersecretion and airway barrier function. Cigarette smoke, being next to environmental pollution the main cause of COPD, is believed to cause epithelial hyperpermeability by disrupting the barrier function. Here we will focus on the most recent progress on compartmentalized signalling by cAMP. In addition to G protein-coupled receptors, adenylyl cyclases, cAMP-specific phospho-diesterases (PDEs) maintain compartmentalized cAMP signalling. Intriguingly, spatially discrete cAMP-sensing signalling complexes seem also to involve distinct members of the A-kinase anchoring (AKAP) superfamily and IQ motif containing GTPase activating protein (IQGAPs). In this review, we will highlight the interaction between cAMP and the epithelial barrier to retain proper lung function and to alleviate COPD symptoms and focus on the possible molecular mechanisms involved in this process. Future studies should include the development of cAMP-sensing multiprotein complex specific disruptors and/or stabilizers to orchestrate cellular functions. Compartmentalized cAMP signalling regulates important cellular processes in the lung and may serve as a therapeutic target.
Collapse
Affiliation(s)
- Anouk Oldenburger
- Department of Molecular Pharmacology, Groningen Research Institute for Pharmacy, University of Groningen, 9713 AV, Groningen, The Netherlands.
| | | | | |
Collapse
|
24
|
Smith JP, Uhernik AL, Li L, Liu Z, Drewes LR. Regulation of Mct1 by cAMP-dependent internalization in rat brain endothelial cells. Brain Res 2012; 1480:1-11. [PMID: 22925948 DOI: 10.1016/j.brainres.2012.08.026] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2012] [Revised: 08/08/2012] [Accepted: 08/15/2012] [Indexed: 10/28/2022]
Abstract
In the cerebrovascular endothelium, monocarboxylic acid transporter 1 (Mct1) controls blood-brain transport of short chain monocarboxylic and keto acids, including pyruvate and lactate, to support brain energy metabolism. Mct1 function is acutely decreased in rat brain cerebrovascular endothelial cells by β-adrenergic signaling through cyclic adenosine monophosphate (cAMP); however, the mechanism for this acute reduction in transport capacity is unknown. In this report, we demonstrate that cAMP induces the dephosphorylation and internalization of Mct1 from the plasma membrane into caveolae and early endosomes in the RBE4 rat brain cerebrovascular endothelial cell line. Additionally, we provide evidence that Mct1 constitutively cycles through clathrin vesicles and recycling endosomes in a pathway that is not dependent upon cAMP signaling in these cells. Our results are important because they show for the first time the regulated and unregulated vesicular trafficking of Mct1 in cerebrovascular endothelial cells; processes which have significance for better understanding normal brain energy metabolism, and the etiology and potential therapeutic approaches to treating brain diseases, such as stroke, in which lactic acidosis is a key component.
Collapse
Affiliation(s)
- Jeffrey P Smith
- Colorado State University-Pueblo, Department of Biology, 2200 Bonforte Blvd., Pueblo, CO 81001, USA.
| | | | | | | | | |
Collapse
|
25
|
Mi Q, Chen N, Shaifta Y, Xie L, Lu H, Liu Z, Chen Q, Hamid C, Becker S, Ji Y, Ferro A. Activation of endothelial nitric oxide synthase is dependent on its interaction with globular actin in human umbilical vein endothelial cells. J Mol Cell Cardiol 2011; 51:419-27. [PMID: 21741389 DOI: 10.1016/j.yjmcc.2011.06.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2011] [Revised: 06/01/2011] [Accepted: 06/21/2011] [Indexed: 10/24/2022]
Abstract
Endothelial nitric oxide synthase (eNOS) has been reported to associate with globular actin, and this association increases eNOS activity. Adenosine, histamine, salbutamol and thrombin cause activation of eNOS through widely different mechanisms. Whether these eNOS agonists can regulate eNOS activity through affecting its association with actin is unknown. As previously reported, we confirmed in cultured human umbilical vein endothelial cells (HUVEC) that histamine and thrombin increased intracellular Ca(2+) whereas adenosine and salbutamol did not, and that these four agonists caused different effects on actin filament structure. Nevertheless, despite their divergent effects on intracellular Ca(2+) and on actin filament structure, we found by immunoprecipitation that adenosine, histamine, salbutamol and thrombin all caused an increase in association between eNOS and globular actin. This increase of association was inhibited by pre-treatment with phalloidin, an actin filament stabilizer. All of these agonists also increased phosphorylation of eNOS on serine residue 1177, eNOS activity, and cyclic guanosine-3', 5'-monophosphate, and these increases were all attenuated by phalloidin. Agonist-induced phosphorylation of eNOS on serine 1177 was attenuated by Akt inhibition, whereas association of eNOS with actin was not. We also found, in HEK-293 cells transfected with the eNOS mutants eNOS-S1177A or eNOS-S1177D, that the association between eNOS and globular actin was decreased as compared to cells transfected with wild-type eNOS. We conclude that association of globular actin with eNOS plays an essential and necessary role in agonist-induced eNOS activation, through enabling its phosphorylation by Akt at serine residue 1177.
Collapse
Affiliation(s)
- Qiongyu Mi
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Key Laboratory of Human Functional Genomics, Nanjing Medical University, Nanjing, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Sayner SL. Emerging themes of cAMP regulation of the pulmonary endothelial barrier. Am J Physiol Lung Cell Mol Physiol 2011; 300:L667-78. [PMID: 21335524 DOI: 10.1152/ajplung.00433.2010] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The presence of excess fluid in the interstitium and air spaces of the lung presents severe restrictions to gas exchange. The pulmonary endothelial barrier regulates the flux of fluid and plasma proteins from the vascular space into the underlying tissue. The integrity of this endothelial barrier is dynamically regulated by transitions in cAMP (3',5'-cyclic adenosine monophosphate), which are synthesized in discrete subcellular compartments. Cyclic AMP generated in the subplasma membrane compartment acts through PKA and Epac (exchange protein directly activated by cAMP) to tighten cell adhesions, strengthen cortical actin, reduce actomyosin contraction, and decrease permeability. Confining cAMP within the subplasma membrane space is critical to its barrier-protective properties. When cAMP escapes the near membrane compartment and gains access to the cytosolic compartment, or when soluble adenylyl cyclases generate cAMP within the cytosolic compartment, this second messenger activates established cytosolic cAMP signaling cascades to perturb the endothelial barrier through PKA-mediated disruption of microtubules. Thus the concept of cAMP compartmentalization in endothelial barrier regulation is gaining momentum and new possibilities are being unveiled for cytosolic cAMP signaling with the emergence of the bicarbonate-regulated mammalian soluble adenylyl cyclase (sAC or AC10).
Collapse
Affiliation(s)
- Sarah L Sayner
- Dept. of Cell Biology and Neuroscience, Member, Center for Lung Biology, College of Medicine, Univ. of South Alabama, Mobile, AL 36688, USA.
| |
Collapse
|
27
|
Liu F, Mih JD, Shea BS, Kho AT, Sharif AS, Tager AM, Tschumperlin DJ. Feedback amplification of fibrosis through matrix stiffening and COX-2 suppression. ACTA ACUST UNITED AC 2010; 190:693-706. [PMID: 20733059 PMCID: PMC2928007 DOI: 10.1083/jcb.201004082] [Citation(s) in RCA: 582] [Impact Index Per Article: 41.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Tissue stiffening is a hallmark of fibrotic disorders but has traditionally been regarded as an outcome of fibrosis, not a contributing factor to pathogenesis. In this study, we show that fibrosis induced by bleomycin injury in the murine lung locally increases median tissue stiffness sixfold relative to normal lung parenchyma. Across this pathophysiological stiffness range, cultured lung fibroblasts transition from a surprisingly quiescent state to progressive increases in proliferation and matrix synthesis, accompanied by coordinated decreases in matrix proteolytic gene expression. Increasing matrix stiffness strongly suppresses fibroblast expression of COX-2 (cyclooxygenase-2) and synthesis of prostaglandin E(2) (PGE(2)), an autocrine inhibitor of fibrogenesis. Exogenous PGE(2) or an agonist of the prostanoid EP2 receptor completely counteracts the proliferative and matrix synthetic effects caused by increased stiffness. Together, these results demonstrate a dominant role for normal tissue compliance, acting in part through autocrine PGE(2), in maintaining fibroblast quiescence and reveal a feedback relationship between matrix stiffening, COX-2 suppression, and fibroblast activation that promotes and amplifies progressive fibrosis.
Collapse
Affiliation(s)
- Fei Liu
- Molecular and Integrative Physiological Sciences, Department of Environmental Health, Harvard School of Public Health, Boston, MA 02115
| | | | | | | | | | | | | |
Collapse
|
28
|
Wong KHK, Truslow JG, Tien J. The role of cyclic AMP in normalizing the function of engineered human blood microvessels in microfluidic collagen gels. Biomaterials 2010; 31:4706-14. [PMID: 20303168 DOI: 10.1016/j.biomaterials.2010.02.041] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2009] [Accepted: 02/16/2010] [Indexed: 11/29/2022]
Abstract
Nearly all engineered tissues must eventually be vascularized to survive. To this end, we and others have recently developed methods to synthesize extracellular matrix-based scaffolds that contain open microfluidic networks. These scaffolds serve as templates for the formation of endothelial tubes that can be perfused; whether such microvascular structures are stable and/or functional is largely unknown. Here, we show that compounds that elevate intracellular concentrations of the second messenger cyclic AMP (cAMP) strongly normalize the phenotype of engineered human microvessels in microfluidic type I collagen gels. Cyclic AMP-elevating agents promoted vascular stability and barrier function, and reduced cellular turnover. Under conditions that induced the highest levels of cAMP, the physiology of engineered microvessels in vitro quantitatively mirrored that of native vessels in vivo. Computational analysis indicated that cAMP stabilized vessels partly via its enhancement of barrier function.
Collapse
Affiliation(s)
- Keith H K Wong
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | | | | |
Collapse
|
29
|
Aslam M, Härtel FV, Arshad M, Gündüz D, Abdallah Y, Sauer H, Piper HM, Noll T. cAMP/PKA antagonizes thrombin-induced inactivation of endothelial myosin light chain phosphatase: role of CPI-17. Cardiovasc Res 2010; 87:375-84. [PMID: 20202976 DOI: 10.1093/cvr/cvq065] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
AIMS Activation of cAMP signalling abrogates thrombin-induced hyperpermeability. One of the mechanisms underlying this protective effect is the inactivation of endothelial contractile machinery, one of the major determinants of endothelial barrier function, mainly via the activation of myosin light chain phosphatase (MLCP). To date, the mechanisms of cAMP-mediated MLCP activation are only partially understood. Here the contribution of two cAMP effectors, PKA and Epac, in the regulation of endothelial contractile machinery and barrier function was studied. METHODS AND RESULTS Endothelial contractile machinery and barrier function were analysed in cultured human umbilical vein endothelial cells (HUVEC). The cAMP analogues 8-CPT-cAMP and 6-Bnz-cAMP were used to activate Epac and PKA, respectively, and forskolin (FSK) was used to activate adenylyl cyclase. The cells were challenged by thrombin to inhibit MLCP via the RhoA/Rock pathway. Activation of either PKA or Epac partially blocked thrombin-induced hyperpermeability. Simultaneous activation of PKA and Epac had additive effects that were comparable to that of FSK. Activation of PKA but not Epac inhibited thrombin-induced phosphorylation of MLC and the MLCP regulatory subunit MYPT1, partly via inhibition of the RhoA/Rock pathway. FSK activated the MLCP catalytic subunit PP1 via dephosphorylation and dissociation of the PP1 inhibitory protein CPI-17. FSK blunted thrombin-induced CPI-17 phosphorylation, CPI-17/PP1 complex formation, and PP1 inactivation. Down-regulation of CPI-17 attenuated thrombin-induced hyperpermeability and abolished the antagonistic effect of the PKA activator, whereas the Epac activator retained its antagonistic effect. CONCLUSION cAMP/PKA regulates the endothelial barrier via inhibition of the contractile machinery, mainly by the activation of MLCP via inhibition of CPI-17 and RhoA/Rock. The permeability-lowering effect of the cAMP/Epac pathway is independent of CPI-17.
Collapse
Affiliation(s)
- Muhammad Aslam
- Physiologisches Institut, Justus-Liebig-Universität, Aulweg 129, Giessen, Germany
| | | | | | | | | | | | | | | |
Collapse
|
30
|
ADF/cofilin: a functional node in cell biology. Trends Cell Biol 2010; 20:187-95. [PMID: 20133134 DOI: 10.1016/j.tcb.2010.01.001] [Citation(s) in RCA: 549] [Impact Index Per Article: 39.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2009] [Revised: 12/22/2009] [Accepted: 01/05/2010] [Indexed: 12/12/2022]
Abstract
Recent findings have significantly expanded our understanding of the regulation of actin-depolymerizing factor (ADF)/cofilin proteins and the profound multifaceted impact that these well-established regulators of actin dynamics have on cell biology. In this review we discuss new aspects of previously documented regulation, such as phosphorylation, but also cover novel recently established modes of regulation and functions of ADF (also known as destrin)/cofilin. We now understand that their activity responds to a vast array of inputs far greater than previously appreciated and that these proteins not only feed back to the crucially important dynamics of actin, but also to apoptosis cascades, phospholipid metabolism, and gene expression. We argue that this ability to respond to physiological changes by modulating those same changes makes the ADF/cofilin protein family a homeostatic regulator or 'functional node' in cell biology.
Collapse
|
31
|
Prasain N, Alexeyev M, Balczon R, Stevens T. Soluble adenylyl cyclase-dependent microtubule disassembly reveals a novel mechanism of endothelial cell retraction. Am J Physiol Lung Cell Mol Physiol 2009; 297:L73-83. [PMID: 19395666 DOI: 10.1152/ajplung.90577.2008] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Soluble adenylyl cyclase toxins, such as Pseudomonas aeruginosa exoY, generate a cAMP pool that retracts cell borders. However, the cytoskeletal basis by which this cAMP signal retracts cell borders is not known. We sought to determine whether activation of chimeric, soluble adenylyl cyclase I/II (sACI/II) reorganizes either microtubules or peripheral actin. Endothelial cells were stably transfected with either green fluorescent protein-labeled alpha-tubulin or beta-actin, and then infected with adenovirus to express sACI/II. Forskolin, which stimulates both the endogenously expressed transmembrane adenylyl cyclases and sACI/II, induced cell retraction accompanied by the reorganization of peripheral microtubules. However, cortical filamentous-actin (f-actin) did not reorganize into stress fibers, and myosin light-chain-20 phosphorylation was decreased. Isoproterenol, which activates endogenous adenylyl cyclases but does not activate sACI/II, did not induce endothelial cell gaps and did not influence microtubule or f-actin architecture. Thus, sACI/II generates a cAMP signal that reorganizes microtubules and induces cell retraction, without inducing f-actin stress fibers. These findings illustrate that endothelial cell gap formation can proceed without f-actin stress fiber formation, and provide mechanistic insight how bacterial adenylyl cyclase toxins reorganize the cytoskeleton to induce cell rounding.
Collapse
Affiliation(s)
- Nutan Prasain
- Departments of Pharmacology, Center for Lung Biology, University of South Alabama, Mobile, Alabama 36688, USA
| | | | | | | |
Collapse
|
32
|
Jalimarada SS, Shivanna M, Kini V, Mehta D, Srinivas SP. Microtubule disassembly breaks down the barrier integrity of corneal endothelium. Exp Eye Res 2009; 89:333-43. [PMID: 19345211 DOI: 10.1016/j.exer.2009.03.019] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2009] [Revised: 03/01/2009] [Accepted: 03/25/2009] [Indexed: 01/28/2023]
Abstract
Increased contractility of the peri-junctional actomyosin ring (PAMR) breaks down the barrier integrity of corneal endothelium. This study has examined the effects of microtubule disassembly on Myosin Light Chain (MLC) phosphorylation, a biochemical marker of actomyosin contraction, and barrier integrity in monolayers of cultured bovine corneal endothelial cells (BCEC). Exposure to nocodazole, which readily induced microtubule disassembly, led to disruption of the characteristically dense assembly of cortical actin cytoskeleton at the apical junctional complex (i.e., PAMR) and dispersion of ZO-1 from its normal locus. Nocodazole also led to an increase in phosphorylation of MLC. Concomitant with these changes, nocodazole caused an increase in permeability to HRP and FITC dextran (10 kDa) and a decrease in trans-endothelial electrical resistance (TER). Y-27632 (a Rho kinase inhibitor) and forskolin (known to inhibit activation of RhoA through direct elevation of cAMP) opposed the nocodazole-induced MLC phosphorylation, decrease in TER, and dispersion of ZO-1. Thrombin, which breaks down the barrier integrity of BCEC monolayers, also induced microtubule disassembly and MLC phosphorylation. Pre-treatment with paclitaxel to stabilize microtubules opposed the thrombin effects. These results suggest that microtubule disassembly breaks down the barrier integrity of BCEC through activation of RhoA and subsequent disruption of the PAMR. The thrombin effect also highlights that signaling downstream of GPCRs can also influence the organization of microtubules.
Collapse
|
33
|
Ihnatovych I, Livak M, Reed J, de Lanerolle P, Strakova Z. Manipulating actin dynamics affects human in vitro decidualization. Biol Reprod 2009; 81:222-30. [PMID: 19339710 DOI: 10.1095/biolreprod.108.074666] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The differentiation of uterine stromal fibroblasts into decidual cells is critical for establishing pregnancy. This process, called decidualization, requires the reorganization of the actin cytoskeleton, which mainly depends on actin dynamics and the phosphorylation status of the myosin light chain. We manipulated actin dynamics with jasplakinolide (100 nM) and latrunculin B (1 microM), both of which significantly inhibited the synthesis of decidualization markers induced by 6 days of treatment with embryo-mimicking stimulus interleukin 1beta (IL1B) and steroid hormones (SHs; 17beta-estradiol and medroxyprogesterone acetate) in the human uterine fibroblast (HuF) in vitro model. However, only jasplakinolide had long-lasting effects on the G-actin:F-actin ratio and prevented decidualization induced by the artificial stimulus cAMP (and SHs). Actin-binding protein cofilin mainly colocalized with G-actin in the nucleus as well as the cytoplasm. Only some spots of colocalization between cofilin and F-actin were detected in the cytoplasm. Brief extraction of cytosolic proteins from living cells revealed that in cells treated with IL1B or cAMP (and SHs) for 6 days, cofilin was mainly detected in the nucleus. The translocation of cofilin from cytosol to nucleus was also detected in HuFs treated for 12 days with SHs, IL1B and SHs, and cAMP and SHs. The same significant translocation was confirmed in primary baboon stromal uterine fibroblasts. We conclude that changes in actin dynamics, particularly the stabilization of F-actin, have a significant negative impact on decidualization, and the translocation of cofilin to the nucleus is a key feature of this process in the primate.
Collapse
Affiliation(s)
- Ivanna Ihnatovych
- Department of Obstetrics and Gynecology, University of Illinois at Chicago, Chicago, Illinois 60612-7313, USA
| | | | | | | | | |
Collapse
|
34
|
|
35
|
Gavard J, Gutkind JS. Protein kinase C-related kinase and ROCK are required for thrombin-induced endothelial cell permeability downstream from Galpha12/13 and Galpha11/q. J Biol Chem 2008; 283:29888-96. [PMID: 18713748 DOI: 10.1074/jbc.m803880200] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Increase in vascular permeability occurs under many physiological conditions such as wound repair, inflammation, and thrombotic reactions and is central in diverse human pathologies, including tumor-induced angiogenesis, ocular diseases, and septic shock. Thrombin is a pro-coagulant serine protease, which causes the local loss of endothelial barrier integrity thereby enabling the rapid extravasation of plasma proteins and the local formation of fibrin-containing clots. Available information suggests that thrombin induces endothelial permeability by promoting actomyosin contractility through the Rho/ROCK signaling pathway. Here we took advantage of pharmacological inhibitors, knockdown approaches, and the emerging knowledge on how permeability factors affect endothelial junctions to investigate in detail the mechanism underlying thrombin-induced endothelial permeability. We show that thrombin signals through PAR-1 and its coupled G proteins Galpha(12/13) and Galpha(11/q) to induce RhoA activation and intracellular calcium elevation, and that these events are interrelated. In turn, this leads to the stimulation of ROCK, which causes actin stress-fiber formation. However, this alone is not sufficient to account for thrombin-induced permeability. Instead, we found that protein kinase C-related kinase, a Rho-dependent serine/threonine kinase, is activated in endothelial cells upon thrombin stimulation and that its expression is required for endothelial permeability and the remodeling of cell-extracellular matrix and cell-cell adhesions. Our results demonstrate that the signal initiated by thrombin bifurcates at the level of RhoA to promote changes in the cytoskeletal architecture through ROCK, and the remodeling of focal adhesion components through protein kinase C-related kinase. Ultimately, both pathways converge to cause cell-cell junction disruption and provoke vascular leakage.
Collapse
Affiliation(s)
- Julie Gavard
- Oral and Pharyngeal Cancer Branch, NIDCR, National Institutes of Health, Bethesda, Maryland 20892, USA.
| | | |
Collapse
|
36
|
Goeckeler ZM, Bridgman PC, Wysolmerski RB. Nonmuscle myosin II is responsible for maintaining endothelial cell basal tone and stress fiber integrity. Am J Physiol Cell Physiol 2008; 295:C994-1006. [PMID: 18701651 DOI: 10.1152/ajpcell.00318.2008] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Cultured confluent endothelial cells exhibit stable basal isometric tone associated with constitutive myosin II regulatory light chain (RLC) phosphorylation. Thrombin treatment causes a rapid increase in isometric tension concomitant with myosin II RLC phosphorylation, actin polymerization, and stress fiber reorganization while inhibitors of myosin light chain kinase (MLCK) and Rho-kinase prevent these responses. These findings suggest a central role for myosin II in the regulation of endothelial cell tension. The present studies examine the effects of blebbistatin, a specific inhibitor of myosin II activity, on basal tone and thrombin-induced tension development. Although blebbistatin treatment abolished basal tension, this was accompanied by an increase in myosin II RLC phosphorylation. The increase in RLC phosphorylation was Ca(2+) dependent and mediated by MLCK. Similarly, blebbistatin inhibited thrombin-induced tension without interfering with the increase in RLC phosphorylation or in F-actin polymerization. Blebbistatin did prevent myosin II filament incorporation and association with polymerizing or reorganized actin filaments leading to the disappearance of stress fibers. Thus the inhibitory effects of blebbistatin on basal tone and induced tension are consistent with a requirement for myosin II activity to maintain stress fiber integrity.
Collapse
Affiliation(s)
- Zoe M Goeckeler
- Department of Neurobiology and Anatomy, West Virginia University School of Medicine, Morgantown, West Virginia 26506, USA
| | | | | |
Collapse
|
37
|
Ramachandran C, Satpathy M, Mehta D, Srinivas SP. Forskolin induces myosin light chain dephosphorylation in bovine trabecular meshwork cells. Curr Eye Res 2008; 33:169-76. [PMID: 18293188 DOI: 10.1080/02713680701837067] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
PURPOSE Enhanced contractility of the actin cytoskeleton in trabecular meshwork (TM) cells is implicated in increased resistance to aqueous humor outflow. In this study, we have investigated effects of forskolin, which is known to elevate cAMP and also enhance aqueous humor outflow, on myosin light chain (MLC) phosphorylation, a biochemical marker of actin contractility. METHODS Experiments were performed using cultured bovine TM cells. Phosphorylated MLC (pMLC), expressed as the % of untreated cells, was assessed by urea-glycerol gel electrophoresis and Western blotting. RhoA activity was determined by affinity precipitation of RhoA-GTP to RhoA binding domain of an effector of RhoA. Intracellular cAMP levels were measured by ELISA. RESULTS Exposure to LPA (lysophosphatidic acid) led to increased MLC phosphorylation (LPA: pMLC=133%) and activation of RhoA. These responses of LPA were suppressed by co-treatment with forskolin (LPA+forskolin: pMLC=88%). Similarly, ET-1 and nocodazole-induced MLC phosphorylation (ET-1: pMLC=145%; nocodazole: pMLC=145%) as well as RhoA activation were suppressed by co-treatment with forskolin (ET-1+forskolin: pMLC=99%; nocodazole+forskolin: pMLC=107%). Exposure to forskolin alone led to MLC dephosphorylation (pMLC=68%). Forskolin alone led to a 4-fold increase in cAMP levels. This increase was not affected when co-treated with LPA or ET-1. CONCLUSIONS Forskolin prevents MLC phosphorylation induced by LPA, ET-1, and nocodazole through inhibition of RhoA-Rho kinase axis. MLC dephosphorylation and consequent relaxation of actin cytoskeleton in TM cells presumably underlies the increased outflow facility reported in response to forskolin.
Collapse
|
38
|
Zhu B, Fukada K, Zhu H, Kyprianou N. Prohibitin and cofilin are intracellular effectors of transforming growth factor beta signaling in human prostate cancer cells. Cancer Res 2007; 66:8640-7. [PMID: 16951178 DOI: 10.1158/0008-5472.can-06-1443] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
A proteomic analysis was pursued to identify new signaling effectors of transforming growth factor beta1 (TGF-beta1) that serve as potential intracellular effectors of its apoptotic action in human prostate cancer cells. The androgen-sensitive and TGF-beta-responsive human prostate cancer cells, LNCaP T beta RII, were used as in vitro model. In response to TGF-beta, significant posttranslational changes in two proteins temporally preceded apoptotic cell death. TGF-beta mediated the nuclear export of prohibitin, a protein involved in androgen-regulated prostate growth, to the cytosol in the LNCaP T beta RII cells. Cofilin, a protein involved in actin depolymerization, cell motility, and apoptosis, was found to undergo mitochondrial translocation in response to TGF-beta before cytochrome c release. Loss-of-function approaches (small interfering RNA) to silence prohibitin expression revealed a modest decrease in the apoptotic response to TGF-beta and a significant suppression in TGF-beta-induced cell migration. Silencing Smad4 showed that the cellular localization changes associated with prohibitin and cofilin action in response to TGF-beta are independent of Smad4 intracellular signaling.
Collapse
Affiliation(s)
- Beibei Zhu
- Division of Urology, College of Medicine, University of Kentucky, Lexington, KY 40536-0298, USA
| | | | | | | |
Collapse
|
39
|
Chen X, Zheng Y, Shen Y. Natural Products with Maleic Anhydride Structure: Nonadrides, Tautomycin, Chaetomellic Anhydride, and Other Compounds. Chem Rev 2007; 107:1777-830. [PMID: 17439289 DOI: 10.1021/cr050029r] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Xiaolong Chen
- Institute of Bioengineering, Zhejiang University of Technology, Hangzhou 310032, P. R. China
| | | | | |
Collapse
|
40
|
Qiu J, Gao HQ, Zhou RH, Liang Y, Zhang XH, Wang XP, You BA, Cheng M. Proteomics analysis of the proliferative effect of low-dose ouabain on human endothelial cells. Biol Pharm Bull 2007; 30:247-53. [PMID: 17268060 DOI: 10.1248/bpb.30.247] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Digitalis has been used to treat congestive heart failure for more than 200 years, although the dual effects (proliferation and death) induced by digitalis on cell growth have been known for many years, the mechanisms by which digitalis causes the actions were not completely known. The aim of this work was to characterize the proliferative effect of ouabain on cell growth in endothelial cells, and, to do the differential proteomic analysis of human umbilical vein endothelial cells (HUVEC) in response to ouabain and examine changes in protein expression. HUVEC were exposed to different concentrations (0.1-100 nM) of ouabain at 12-48 h intervals. Cell growth and morphological changes of HUVEC treated with ouabain were compared with cells under nontreated conditions. Ouabain stimulated HUVEC cell proliferation at low concentrations and induced cell death at higher concentrations. Using proteomics study, we identified 32 proteins of HUVEC with various important cellular functions and revealed 8 proteins such as Annexin A1, Annexin A2, Malate dehydrogenase, Myosin regulatory light chain 2 (MRLC2), Profilin-1, S100 calcium-binding protein A13, Triosephosphate isomerase and Translationally controlled tumor protein, regulated by low-dose ouabain treatment and MRLC2 was subsequently confirmed by Western blot. Our results give new insights into the cellular and molecular mechanisms of the proliferation action of low-dose ouabain on HUVEC and provide new avenues for the treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Jie Qiu
- Department of Geriatrics, Shandong University Qilu Hospital, Jinan, China
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Lee SY, Kim YT, Lee MS, Kim YB, Chung E, Kim S, Lee JW. Focal adhesion and actin organization by a cross-talk of TM4SF5 with integrin alpha2 are regulated by serum treatment. Exp Cell Res 2006; 312:2983-99. [PMID: 16828471 DOI: 10.1016/j.yexcr.2006.06.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2006] [Revised: 05/18/2006] [Accepted: 06/02/2006] [Indexed: 11/29/2022]
Abstract
The biological functions of transmembrane 4 L6 family member 5 (TM4SF5) homologues to a tumor-associated antigen L6 are unknown, although it is over-expressed in certain forms of cancer. In the present study, the ectopic expression of TM4SF5 in Cos7 cells reduced integrin signaling under serum-containing conditions, but increased integrin signaling upon serum-free replating on substrates. TM4SF5 regulated actin organization and focal contact dynamics via the serum treatment-dependent differential regulation of FAK Tyr925 and paxillin Tyr118 phosphorylations and their localizations on peripheral cell boundaries. Y925F FAK mutation abolished the TM4SF5 effects. TM4SF5 associated with integrin alpha2 subunit, and this association was abolished by serum treatment. Furthermore, functional blocking anti-integrin alpha2 antibody abolished TM4SF5-enhanced signaling activity and caused membrane blebbing with abnormal actin organization. TM4SF5 increased chemotactic but decreased haptotactic migration. Altogether, this study reveals the functions of TM4SF5 collaborative with integrin signaling to alter focal contact dynamics, actin reorganization, and migration. Furthermore, this study suggests a mechanism of cross-talk between TM4SF5 and integrin which is further regulated by growth factor signaling.
Collapse
Affiliation(s)
- Sung-Yul Lee
- Cancer Research Institute, Department of Molecular and Clinical Oncology, College of Medicine, Seoul National University, 28, Yeongeon-dong, Jongno-gu, Seoul 110-799, Korea
| | | | | | | | | | | | | |
Collapse
|
42
|
Lubomirov LT, Reimann K, Metzler D, Hasse V, Stehle R, Ito M, Hartshorne DJ, Gagov H, Pfitzer G, Schubert R. Urocortin-induced decrease in Ca2+ sensitivity of contraction in mouse tail arteries is attributable to cAMP-dependent dephosphorylation of MYPT1 and activation of myosin light chain phosphatase. Circ Res 2006; 98:1159-67. [PMID: 16574904 DOI: 10.1161/01.res.0000219904.43852.3e] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Urocortin, a vasodilatory peptide related to corticotropin-releasing factor, may be an endogenous regulator of blood pressure. In vitro, rat tail arteries are relaxed by urocortin by a cAMP-mediated decrease in myofilament Ca2+ sensitivity through a still unclear mechanism. Here we show that contraction of intact mouse tail arteries induced with 42 mmol/L KCl or 0.5 micromol/L noradrenaline was associated with a approximately 2-fold increase in the phosphorylation of the regulatory subunit of myosin phosphatase (SMPP-1M), MYPT1, at Thr696, which was reversed in arteries relaxed with urocortin. Submaximally (pCa 6.1) contracted mouse tail arteries permeabilized with alpha-toxin were relaxed with urocortin by 39+/-3% at constant [Ca2+], which was associated with a decrease in myosin light chain (MLC20Ser19), MYPT1Thr696, and MYPT1Thr850 phosphorylation by 60%, 28%, and 52%, respectively. The Rho-associated kinase (ROK) inhibitor Y-27632 decreased MYPT1 phosphorylation by a similar extent. Inhibition of PP-2A with 3 nmol/L okadaic acid had no effect on MYPT1 phosphorylation, whereas inhibition of PP-1 with 3 micromol/L okadaic acid prevented dephosphorylation. Urocortin increased the rate of dephosphorylation of MLC20Ser19 approximately 2.2-fold but had no effect on the rate of contraction under conditions of, respectively, inhibited kinase and phosphatase activities. The effect of urocortin on MLC20Ser19 and MYPT1 phosphorylation was blocked by Rp-8-CPT-cAMPS and mimicked by Sp-5,6-DCl-cBIMPS. In summary, these results provide evidence that Ca(2+)-independent relaxation by urocortin can be attributed to a cAMP-mediated increased activity of SMPP-1M which at least in part is attributable to a decrease in the inhibitory phosphorylation of MYPT1.
Collapse
|