1
|
The Molecular Heterogeneity of Store-Operated Ca 2+ Entry in Vascular Endothelial Cells: The Different roles of Orai1 and TRPC1/TRPC4 Channels in the Transition from Ca 2+-Selective to Non-Selective Cation Currents. Int J Mol Sci 2023; 24:ijms24043259. [PMID: 36834672 PMCID: PMC9967124 DOI: 10.3390/ijms24043259] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/31/2023] [Accepted: 02/03/2023] [Indexed: 02/11/2023] Open
Abstract
Store-operated Ca2+ entry (SOCE) is activated in response to the inositol-1,4,5-trisphosphate (InsP3)-dependent depletion of the endoplasmic reticulum (ER) Ca2+ store and represents a ubiquitous mode of Ca2+ influx. In vascular endothelial cells, SOCE regulates a plethora of functions that maintain cardiovascular homeostasis, such as angiogenesis, vascular tone, vascular permeability, platelet aggregation, and monocyte adhesion. The molecular mechanisms responsible for SOCE activation in vascular endothelial cells have engendered a long-lasting controversy. Traditionally, it has been assumed that the endothelial SOCE is mediated by two distinct ion channel signalplexes, i.e., STIM1/Orai1 and STIM1/Transient Receptor Potential Canonical 1(TRPC1)/TRPC4. However, recent evidence has shown that Orai1 can assemble with TRPC1 and TRPC4 to form a non-selective cation channel with intermediate electrophysiological features. Herein, we aim at bringing order to the distinct mechanisms that mediate endothelial SOCE in the vascular tree from multiple species (e.g., human, mouse, rat, and bovine). We propose that three distinct currents can mediate SOCE in vascular endothelial cells: (1) the Ca2+-selective Ca2+-release activated Ca2+ current (ICRAC), which is mediated by STIM1 and Orai1; (2) the store-operated non-selective current (ISOC), which is mediated by STIM1, TRPC1, and TRPC4; and (3) the moderately Ca2+-selective, ICRAC-like current, which is mediated by STIM1, TRPC1, TRPC4, and Orai1.
Collapse
|
2
|
Li D. Role of Spectrin in Endocytosis. Cells 2022; 11:cells11152459. [PMID: 35954302 PMCID: PMC9368487 DOI: 10.3390/cells11152459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/02/2022] [Accepted: 08/06/2022] [Indexed: 11/16/2022] Open
Abstract
Cytoskeletal spectrin is found in (non)erythroid cells. Eukaryotic endocytosis takes place for internalizing cargos from extracellular milieu. The role of spectrin in endocytosis still remains poorly understood. Here, I summarize current knowledge of spectrin function, spectrin-based cytoskeleton and endocytosis of erythrocytes, and highlight how spectrin contributes to endocytosis and working models in different types of cells. From an evolutionary viewpoint, I discuss spectrin and endocytosis in a range of organisms, particularly in plants and yeast where spectrin is absent. Together, the role of spectrin in endocytosis is related to its post-translational modification, movement/rearrangement, elimination (by proteases) and meshwork fencing.
Collapse
Affiliation(s)
- Donghai Li
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, China
| |
Collapse
|
3
|
Mylvaganam S, Riedl M, Vega A, Collins RF, Jaqaman K, Grinstein S, Freeman SA. Stabilization of Endothelial Receptor Arrays by a Polarized Spectrin Cytoskeleton Facilitates Rolling and Adhesion of Leukocytes. Cell Rep 2021; 31:107798. [PMID: 32579925 PMCID: PMC7548125 DOI: 10.1016/j.celrep.2020.107798] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 04/15/2020] [Accepted: 06/01/2020] [Indexed: 12/15/2022] Open
Abstract
Multivalent complexes of endothelial adhesion receptors (e.g., selectins) engage leukocytes to orchestrate their migration to inflamed tissues. Proper anchorage and sufficient density (clustering) of endothelial receptors are required for efficient leukocyte capture and rolling. We demonstrate that a polarized spectrin network dictates the stability of the endothelial cytoskeleton, which is attached to the apical membrane, at least in part, by the abundant transmembrane protein CD44. Single-particle tracking revealed that CD44 undergoes prolonged periods of immobilization as it tethers to the cytoskeleton. The CD44-spectrin "picket fence" alters the behavior of bystander molecules-notably, selectins-curtailing their mobility, inducing their apical accumulation, and favoring their clustering within caveolae. Accordingly, depletion of either spectrin or CD44 virtually eliminated leukocyte rolling and adhesion to the endothelium. Our results indicate that a unique spectrin-based apical cytoskeleton tethered to transmembrane pickets-notably, CD44-is essential for proper extravasation of leukocytes in response to inflammation.
Collapse
Affiliation(s)
- Sivakami Mylvaganam
- Program in Cell Biology, Peter Gilgan Centre for Research and Learning, Hospital for Sick Children, 686 Bay Street, 19-9800, Toronto, ON M5G 0A4, Canada; Department of Biochemistry, University of Toronto, 1 King's College Circle, Toronto, ON M5S 1A8, Canada
| | - Magdalena Riedl
- Program in Cell Biology, Peter Gilgan Centre for Research and Learning, Hospital for Sick Children, 686 Bay Street, 19-9800, Toronto, ON M5G 0A4, Canada
| | - Anthony Vega
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Richard F Collins
- Program in Cell Biology, Peter Gilgan Centre for Research and Learning, Hospital for Sick Children, 686 Bay Street, 19-9800, Toronto, ON M5G 0A4, Canada
| | - Khuloud Jaqaman
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Sergio Grinstein
- Program in Cell Biology, Peter Gilgan Centre for Research and Learning, Hospital for Sick Children, 686 Bay Street, 19-9800, Toronto, ON M5G 0A4, Canada; Department of Biochemistry, University of Toronto, 1 King's College Circle, Toronto, ON M5S 1A8, Canada; Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, ON M5B 1W8, Canada.
| | - Spencer A Freeman
- Program in Cell Biology, Peter Gilgan Centre for Research and Learning, Hospital for Sick Children, 686 Bay Street, 19-9800, Toronto, ON M5G 0A4, Canada; Department of Biochemistry, University of Toronto, 1 King's College Circle, Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
4
|
Srivastava N, Tauseef M, Amin R, Joshi B, Joshi JC, Kini V, Klomp J, Li W, Knezevic N, Barbera N, Siddiqui S, Obukhov A, Karginov A, Levitan I, Komarova Y, Mehta D. Noncanonical function of long myosin light chain kinase in increasing ER-PM junctions and augmentation of SOCE. FASEB J 2020; 34:12805-12819. [PMID: 32772419 PMCID: PMC7496663 DOI: 10.1096/fj.201902462rr] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 06/26/2020] [Accepted: 07/16/2020] [Indexed: 12/13/2022]
Abstract
Increased endothelial permeability leads to excessive exudation of plasma proteins and leukocytes in the interstitium, which characterizes several vascular diseases including acute lung injury. The myosin light chain kinase long (MYLK-L) isoform is canonically known to regulate the endothelial permeability by phosphorylating myosin light chain (MLC-P). Compared to the short MYLK isoform, MYLK-L contains an additional stretch of ~919 amino acid at the N-terminus of unknown function. We show that thapsigargin and thrombin-induced SOCE was markedly reduced in Mylk-L-/- endothelial cells (EC) or MYLK-L-depleted human EC. These agonists also failed to increase endothelial permeability in MYLK-L-depleted EC and Mylk-L-/- lungs, thus demonstrating the novel role of MYLK-L-induced SOCE in increasing vascular permeability. MYLK-L augmented SOCE by increasing endoplasmic reticulum (ER)-plasma membrane (PM) junctions and STIM1 translocation to these junctions. Transduction of N-MYLK domain (amino acids 1-919 devoid of catalytic activity) into Mylk-L-/- EC rescued SOCE to the level seen in control EC in a STIM1-dependent manner. N-MYLK-induced SOCE augmented endothelial permeability without MLC-P via an actin-binding motif, DVRGLL. Liposomal-mediated delivery of N-MYLK mutant but not ∆DVRGLL-N-MYLK mutant in Mylk-L-/- mice rescued vascular permeability increase in response to endotoxin, indicating that targeting of DVRGLL motif within MYLK-L may limit SOCE-induced vascular hyperpermeability.
Collapse
Affiliation(s)
- Nityanand Srivastava
- Department of Pharmacology and Center for Lung and Vascular BiologyThe University of Illinois, College of MedicineChicagoILUSA
| | - Mohammad Tauseef
- Department of Pharmacology and Center for Lung and Vascular BiologyThe University of Illinois, College of MedicineChicagoILUSA
- Department of Pharmaceutical SciencesChicago State University College of PharmacyChicagoILUSA
| | - Ruhul Amin
- Department of Pharmacology and Center for Lung and Vascular BiologyThe University of Illinois, College of MedicineChicagoILUSA
| | - Bhagwati Joshi
- Department of Pharmacology and Center for Lung and Vascular BiologyThe University of Illinois, College of MedicineChicagoILUSA
| | - Jagdish Chandra Joshi
- Department of Pharmacology and Center for Lung and Vascular BiologyThe University of Illinois, College of MedicineChicagoILUSA
| | - Vidisha Kini
- Department of Pharmacology and Center for Lung and Vascular BiologyThe University of Illinois, College of MedicineChicagoILUSA
| | - Jennifer Klomp
- Department of Pharmacology and Center for Lung and Vascular BiologyThe University of Illinois, College of MedicineChicagoILUSA
| | - Weenan Li
- Department of Cellular and Integrative PhysiologyIndiana University School of MedicineIndianapolisINUSA
| | - Nebojsa Knezevic
- Department of Pharmacology and Center for Lung and Vascular BiologyThe University of Illinois, College of MedicineChicagoILUSA
| | - Nicolas Barbera
- Department of MedicineThe Uniiversity of IllinoisChicagoILUSA
| | - Shahid Siddiqui
- Department of Pharmacology and Center for Lung and Vascular BiologyThe University of Illinois, College of MedicineChicagoILUSA
| | - Alexander Obukhov
- Department of Cellular and Integrative PhysiologyIndiana University School of MedicineIndianapolisINUSA
| | - Andrei Karginov
- Department of Pharmacology and Center for Lung and Vascular BiologyThe University of Illinois, College of MedicineChicagoILUSA
| | - Irena Levitan
- Department of MedicineThe Uniiversity of IllinoisChicagoILUSA
| | - Yulia Komarova
- Department of Pharmacology and Center for Lung and Vascular BiologyThe University of Illinois, College of MedicineChicagoILUSA
| | - Dolly Mehta
- Department of Pharmacology and Center for Lung and Vascular BiologyThe University of Illinois, College of MedicineChicagoILUSA
- Department of Pharmaceutical SciencesChicago State University College of PharmacyChicagoILUSA
| |
Collapse
|
5
|
Haldar B, Hamilton CL, Solodushko V, Abney KA, Alexeyev M, Honkanen RE, Scammell JG, Cioffi DL. S100A6 is a positive regulator of PPP5C-FKBP51-dependent regulation of endothelial calcium signaling. FASEB J 2020; 34:3179-3196. [PMID: 31916625 DOI: 10.1096/fj.201901777r] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 10/18/2019] [Accepted: 12/19/2019] [Indexed: 11/11/2022]
Abstract
ISOC is a cation current permeating the ISOC channel. In pulmonary endothelial cells, ISOC activation leads to formation of inter-endothelial cell gaps and barrier disruption. The immunophilin FK506-binding protein 51 (FKBP51), in conjunction with the serine/threonine protein phosphatase 5C (PPP5C), inhibits ISOC . Free PPP5C assumes an autoinhibitory state, which has low "basal" catalytic activity. Several S100 protein family members bind PPP5C increasing PPP5C catalytic activity in vitro. One of these family members, S100A6, exhibits a calcium-dependent translocation to the plasma membrane. The goal of this study was to determine whether S100A6 activates PPP5C in pulmonary endothelial cells and contributes to ISOC inhibition by the PPP5C-FKBP51 axis. We observed that S100A6 activates PPP5C to dephosphorylate tau T231. Following ISOC activation, cytosolic S100A6 translocates to the plasma membrane and interacts with the TRPC4 subunit of the ISOC channel. Global calcium entry and ISOC are decreased by S100A6 in a PPP5C-dependent manner and by FKBP51 in a S100A6-dependent manner. Further, calcium entry-induced endothelial barrier disruption is decreased by S100A6 dependent upon PPP5C, and by FKBP51 dependent upon S100A6. Overall, these data reveal that S100A6 plays a key role in the PPP5C-FKBP51 axis to inhibit ISOC and protect the endothelial barrier against calcium entry-induced disruption.
Collapse
Affiliation(s)
- Barnita Haldar
- Departments of Biochemistry and Molecular Biology, University of South Alabama, Mobile, AL, USA.,Center for Lung Biology, University of South Alabama, Mobile, AL, USA
| | - Caleb L Hamilton
- Department of Anatomy and Molecular Medicine, Alabama College of Osteopathic Medicine, Dothan, AL, USA
| | - Viktoriya Solodushko
- Departments of Biochemistry and Molecular Biology, University of South Alabama, Mobile, AL, USA
| | - Kevin A Abney
- Departments of Biochemistry and Molecular Biology, University of South Alabama, Mobile, AL, USA
| | - Mikhail Alexeyev
- Center for Lung Biology, University of South Alabama, Mobile, AL, USA.,Physiology and Cell Biology, University of South Alabama, Mobile, AL, USA
| | - Richard E Honkanen
- Departments of Biochemistry and Molecular Biology, University of South Alabama, Mobile, AL, USA
| | | | - Donna L Cioffi
- Departments of Biochemistry and Molecular Biology, University of South Alabama, Mobile, AL, USA.,Center for Lung Biology, University of South Alabama, Mobile, AL, USA
| |
Collapse
|
6
|
Goodman SR, Johnson D, Youngentob SL, Kakhniashvili D. The Spectrinome: The Interactome of a Scaffold Protein Creating Nuclear and Cytoplasmic Connectivity and Function. Exp Biol Med (Maywood) 2019; 244:1273-1302. [PMID: 31483159 DOI: 10.1177/1535370219867269] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
We provide a review of Spectrin isoform function in the cytoplasm, the nucleus, the cell surface, and in intracellular signaling. We then discuss the importance of Spectrin’s E2/E3 chimeric ubiquitin conjugating and ligating activity in maintaining cellular homeostasis. Finally we present spectrin isoform subunit specific human diseases. We have created the Spectrinome, from the Human Proteome, Human Reactome and Human Atlas data and demonstrated how it can be a useful tool in visualizing and understanding spectrins myriad of cellular functions.Impact statementSpectrin was for the first 12 years after its discovery thought to be found only in erythrocytes. In 1981, Goodman and colleagues1found that spectrin-like molecules were ubiquitously found in non-erythroid cells leading to a great multitude of publications over the next thirty eight years. The discovery of multiple spectrin isoforms found associated with every cellular compartment, and representing 2-3% of cellular protein, has brought us to today’s understanding that spectrin is a scaffolding protein, with its own E2/E3 chimeric ubiquitin conjugating ligating activity that is involved in virtually every cellular function. We cover the history, localized functions of spectrin isoforms, human diseases caused by mutations, and provide the spectrinome: a useful tool for understanding the myriad of functions for one of the most important proteins in all eukaryotic cells.
Collapse
Affiliation(s)
- Steven R Goodman
- Department of Pediatrics, Memphis Institute of Regenerative Medicine, The University of Tennessee Health Science Center, Memphis, TN 38103
| | - Daniel Johnson
- Department of Pediatrics, Memphis Institute of Regenerative Medicine, The University of Tennessee Health Science Center, Memphis, TN 38103
| | - Steven L Youngentob
- Department of Anatomy and Neurobiology, Memphis Institute of Regenerative Medicine, The University of Tennessee Health Science Center, Memphis, TN 38103
| | - David Kakhniashvili
- Department of Pediatrics, Memphis Institute of Regenerative Medicine, The University of Tennessee Health Science Center, Memphis, TN 38103
| |
Collapse
|
7
|
Zhou C, Francis CM, Xu N, Stevens T. The role of endothelial leak in pulmonary hypertension (2017 Grover Conference Series). Pulm Circ 2018; 8:2045894018798569. [PMID: 30124139 PMCID: PMC6134503 DOI: 10.1177/2045894018798569] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
The canonical transient receptor potential 4 (TRPC4) protein contributes to the molecular make-up of endothelial store-operated calcium entry channels. Store-operated calcium entry is a prominent mode of calcium influx in endothelium. Store-operated calcium entry channels are activated by inflammatory mediators and growth factors, and in endothelium, this process induces inter-endothelial cell gaps that increase permeability. Pulmonary endothelium within extra-alveolar segments, including pulmonary arteries, is especially sensitive to the activation of store-operated calcium entry. Pulmonary arterial hypertension (PAH) is characterized by endothelial cell dysfunction in arteries. As one of the topics for the 2017 Grover Conference Series, we examined whether an endothelial cell permeability defect accompanies PAH and, if so, whether TRPC4 contributes to this defect. Through a series of studies conducted over the past five years, we find endothelial cell barrier dysfunction occurs early in the progression of experimental PAH. Endothelium within the arterial segment, and perhaps in other vascular segments, is highly susceptible to disruption secondary to both activation of store-operated calcium entry channels and high flow. This phenomenon partly depends upon TRPC4 channels. We discuss whether endothelial cell hyperpermeability is relevant to human disease, and more specifically, whether it is relevant to all groups of pulmonary hypertension.
Collapse
Affiliation(s)
- Chun Zhou
- 1 Department of Physiology and Cell Biology, University of South Alabama, Mobile, AL, USA.,2 Center for Lung Biology, University of South Alabama, Mobile, AL, USA
| | - C Michael Francis
- 1 Department of Physiology and Cell Biology, University of South Alabama, Mobile, AL, USA.,2 Center for Lung Biology, University of South Alabama, Mobile, AL, USA
| | - Ningyong Xu
- 1 Department of Physiology and Cell Biology, University of South Alabama, Mobile, AL, USA.,2 Center for Lung Biology, University of South Alabama, Mobile, AL, USA
| | - Troy Stevens
- 1 Department of Physiology and Cell Biology, University of South Alabama, Mobile, AL, USA.,2 Center for Lung Biology, University of South Alabama, Mobile, AL, USA.,3 Department of Internal Medicine, University of South Alabama, Mobile, AL, USA
| |
Collapse
|
8
|
Hamilton CL, Abney KA, Vasauskas AA, Alexeyev M, Li N, Honkanen RE, Scammell JG, Cioffi DL. Serine/threonine phosphatase 5 (PP5C/PPP5C) regulates the ISOC channel through a PP5C-FKBP51 axis. Pulm Circ 2017; 8:2045893217753156. [PMID: 29283027 PMCID: PMC6018905 DOI: 10.1177/2045893217753156] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Pulmonary endothelial cells express a store-operated calcium entry current (Isoc), which contributes to inter-endothelial cell gap formation. Isoc is regulated by a heterocomplex of proteins that includes the immunophilin FKBP51. FKBP51 inhibits Isoc by mechanisms that are not fully understood. In pulmonary artery endothelial cells (PAECs) we have shown that FKBP51 increases microtubule polymerization, an event that is critical for Isoc inhibition by FKBP51. In neurons, FKBP51 promotes microtubule stability through facilitation of tau dephosphorylation. However, FKBP51 does not possess phosphatase activity. Protein phosphatase 5 (PP5C/PPP5C) can dephosphorylate tau, and similar to FKBP51, PP5C possesses tetratricopeptide repeats (TPR) that mediate interaction with heat shock protein-90 (HSP90) chaperone/scaffolding complexes. We therefore tested whether PP5C contributes to FKBP51-mediated inhibition of Isoc. Both siRNA-mediated suppression of PP5C expression in PAECs and genetic disruption of PP5C in HEK293 cells attenuate FKBP51-mediated inhibition of Isoc. Reintroduction of catalytically competent, but not catalytically inactive PP5C, restored FKBP51-mediated inhibition of Isoc. PAEC cell fractionation studies identified both PP5C and the ISOC heterocomplex in the same membrane fractions. Further, PP5C co-precipitates with TRPC4, an essential subunit of ISOC channel. Finally, to determine if PP5C is required for FKBP51-mediated inhibition of calcium entry-induced inter-endothelial cell gap formation, we measured gap area by wide-field microscopy and performed biotin gap quantification assay and electric cell-substrate impedance sensing (ECIS®). Collectively, the data presented indicate that suppression of PP5C expression negates the protective effect of FKBP51. These observations identify PP5C as a novel member of the ISOC heterocomplex that is required for FKBP51-mediated inhibition of Isoc.
Collapse
Affiliation(s)
| | | | | | | | - Ni Li
- University of South Alabama
| | | | | | | |
Collapse
|
9
|
Hamilton CL, Kadeba PI, Vasauskas AA, Solodushko V, McClinton AK, Alexeyev M, Scammell JG, Cioffi DL. Protective role of FKBP51 in calcium entry-induced endothelial barrier disruption. Pulm Circ 2017; 8:2045893217749987. [PMID: 29261039 PMCID: PMC5798693 DOI: 10.1177/2045893217749987] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Pulmonary artery endothelial cells (PAECs) express a cation current, ISOC (store-operated calcium entry current), which when activated permits calcium entry leading to inter-endothelial cell gap formation. The large molecular weight immunophilin FKBP51 inhibits ISOC but not other calcium entry pathways in PAECs. However, it is unknown whether FKBP51-mediated inhibition of ISOC is sufficient to protect the endothelial barrier from calcium entry-induced disruption. The major objective of this study was to determine whether FKBP51-mediated inhibition of ISOC leads to decreased calcium entry-induced inter-endothelial gap formation and thus preservation of the endothelial barrier. Here, we measured the effects of thapsigargin-induced ISOC on the endothelial barrier in control and FKBP51 overexpressing PAECs. FKBP51 overexpression decreased actin stress fiber and inter-endothelial cell gap formation in addition to attenuating the decrease in resistance observed with control cells using electric cell-substrate impedance sensing. Finally, the thapsigargin-induced increase in dextran flux was abolished in FKBP51 overexpressing PAECs. We then measured endothelial permeability in perfused lungs of FKBP51 knockout (FKBP51–/–) mice and observed increased calcium entry-induced permeability compared to wild-type mice. To begin to dissect the mechanism underlying the FKBP51-mediated inhibition of ISOC, a second goal of this study was to determine the role of the microtubule network. We observed that FKBP51 overexpressing PAECs exhibited increased microtubule polymerization that is critical for inhibition of ISOC by FKBP51. Overall, we have identified FKBP51 as a novel regulator of endothelial barrier integrity, and these findings are significant as they reveal a protective mechanism for endothelium against calcium entry-induced disruption.
Collapse
Affiliation(s)
- Caleb L Hamilton
- 1 5557 Department of Biochemistry and Molecular Biology , University of South Alabama, Mobile, AL, USA.,2 Center for Lung Biology, University of South Alabama, Mobile, AL, USA
| | - Pierre I Kadeba
- 1 5557 Department of Biochemistry and Molecular Biology , University of South Alabama, Mobile, AL, USA.,2 Center for Lung Biology, University of South Alabama, Mobile, AL, USA
| | - Audrey A Vasauskas
- 3 376598 Department of Anatomical Sciences and Molecular Medicine , Alabama College of Osteopathic Medicine, Dothan, AL, USA
| | - Viktoriya Solodushko
- 1 5557 Department of Biochemistry and Molecular Biology , University of South Alabama, Mobile, AL, USA
| | - Anna K McClinton
- 2 Center for Lung Biology, University of South Alabama, Mobile, AL, USA.,4 Department of Pharmacology, University of South Alabama, Mobile, AL, USA
| | - Mikhail Alexeyev
- 2 Center for Lung Biology, University of South Alabama, Mobile, AL, USA.,5 Department of Physiology and Cell Biology, University of South Alabama, Mobile, AL, USA
| | - Jonathan G Scammell
- 6 Department of Comparative Medicine, 5557 University of South Alabama , Mobile, AL, USA
| | - Donna L Cioffi
- 1 5557 Department of Biochemistry and Molecular Biology , University of South Alabama, Mobile, AL, USA.,2 Center for Lung Biology, University of South Alabama, Mobile, AL, USA
| |
Collapse
|
10
|
Wu XT, Sun LW, Yang X, Ding D, Han D, Fan YB. The potential role of spectrin network in the mechanotransduction of MLO-Y4 osteocytes. Sci Rep 2017; 7:40940. [PMID: 28112189 PMCID: PMC5256107 DOI: 10.1038/srep40940] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 12/13/2016] [Indexed: 02/06/2023] Open
Abstract
The spectrin is first identified as the main component of erythrocyte membrane skeleton. It is getting growing attention since being found in multiple nonerythroid cells, providing complex mechanical properties and signal interface under the cell membrane. Recent genomics studies have revealed that the spectrin is highly relevant to bone disorders. However, in osteocytes, the important mechanosensors in bone, the role of spectrin is poorly understood. In this research, the role of spectrin in the mechanotransduction of MLO-Y4 osteocytes was studied. Immunofluorescence staining showed that, the spectrins were elaborately organized as a porous network throughout the cytoplasm, and linked with F-actin into a dense layer underlying the cell membrane. AFM results indicate that, the spectrin is pivotal for maintaining the overall elasticity of osteocytes, especially for the cell cortex stiffiness. Disruption of the spectrin network caused obvious softening of osteocytes, and resulted in a significant increase of Ca2+ influx, NO secretion, cell-cell connections and also induced a translocation of eNOS from membrane to cytoplasm. These results indicate that the spectrin network is a global structural support for osteocytes involving in the mechanotransduction process, making it a potential therapeutic target for bone disorders.
Collapse
Affiliation(s)
- Xin-Tong Wu
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, 37th Xue-yuan Road, Hian-dian District, Beijing, China
| | - Lian-Wen Sun
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, 37th Xue-yuan Road, Hian-dian District, Beijing, China.,International Joint Research Center of Aerospace Biotechnology and Medical Engineering, Ministry of Science and Technology of China, School of Biological Science and Medical Engineering, Beihang University, 37th Xue-yuan Road, Hian-dian District, Beijing, China
| | - Xiao Yang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, 37th Xue-yuan Road, Hian-dian District, Beijing, China
| | - Dong Ding
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, 37th Xue-yuan Road, Hian-dian District, Beijing, China
| | - Dong Han
- National Center for Nanoscience and Technology, Beijing, China
| | - Yu-Bo Fan
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, 37th Xue-yuan Road, Hian-dian District, Beijing, China.,National Research Center for Rehabilitation Technical Aids, 1th Ronghuazhong Road, Beijing Economic and Technological Development Zone, China
| |
Collapse
|
11
|
Tam JCW, Ko CH, Koon CM, Cheng Z, Lok WH, Lau CP, Leung PC, Fung KP, Chan WY, Lau CBS. Identification of Target Genes Involved in Wound Healing Angiogenesis of Endothelial Cells with the Treatment of a Chinese 2-Herb Formula. PLoS One 2015; 10:e0139342. [PMID: 26430762 PMCID: PMC4591983 DOI: 10.1371/journal.pone.0139342] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Accepted: 09/11/2015] [Indexed: 01/10/2023] Open
Abstract
Angiogenesis is vitally important in diabetic wound healing. We had previously demonstrated that a Chinese 2-herb formula (NF3) significantly stimulated angiogenesis of HUVEC in wound healing. However, the molecular mechanism has not yet been elucidated. In line with this, global expression profiling of NF3-treated HUVEC was performed so as to assess the regulatory role of NF3 involved in the underlying signaling pathways in wound healing angiogenesis. The microarray results illustrated that different panels of differentially expressed genes were strictly governed in NF3-treated HUVEC in a time-regulated manner. The microarray analysis followed by qRT-PCR and western blotting verification of NF3-treated HUVEC at 6 h revealed the involvement of various genes in diverse biological process, e.g., MAP3K14 in anti-inflammation; SLC5A8 in anti-tumorogenesis; DNAJB7 in protein translation; BIRC5, EPCAM, INSL4, MMP8 and NPR3 in cell proliferation; CXCR7, EPCAM, HAND1 and MMP8 in migration; CXCR7, EPCAM and MMP8 in tubular formation; and BIRC5, CXCR7, EPCAM, HAND1, MMP8 and UBD in angiogenesis. After 16 h incubation of NF3, other sets of genes were shown with differential expression in HUVEC, e.g., IL1RAPL2 and NR1H4 in anti-inflammation; miR28 in anti-tumorogenesis; GRIN1 and LCN1 in anti-oxidation; EPB41 in intracellular signal transduction; PRL and TFAP2A in cell proliferation; miR28, PRL and SCG2 in cell migration; PRL in tubular formation; and miR28, NR1H4 and PRL in angiogenesis. This study provided concrete scientific evidence in support of the regulatory role of NF3 on endothelial cells involved in wound healing angiogenesis.
Collapse
Affiliation(s)
- Jacqueline Chor Wing Tam
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
- State Key Laboratory of Phytochemistry and Plant Resources in West China, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| | - Chun Hay Ko
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
- State Key Laboratory of Phytochemistry and Plant Resources in West China, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| | - Chi Man Koon
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
- State Key Laboratory of Phytochemistry and Plant Resources in West China, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| | - Zhang Cheng
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
- State Key Laboratory of Phytochemistry and Plant Resources in West China, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| | - Wong Hing Lok
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
- State Key Laboratory of Phytochemistry and Plant Resources in West China, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| | - Ching Po Lau
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
- State Key Laboratory of Phytochemistry and Plant Resources in West China, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| | - Ping Chung Leung
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
- State Key Laboratory of Phytochemistry and Plant Resources in West China, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| | - Kwok Pui Fung
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
- State Key Laboratory of Phytochemistry and Plant Resources in West China, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| | - Wai Yee Chan
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| | - Clara Bik San Lau
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
- State Key Laboratory of Phytochemistry and Plant Resources in West China, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
- * E-mail:
| |
Collapse
|
12
|
Abstract
In this minireview, we cover the discovery of the human erythrocyte α spectrin E2/E3 ubiquitin conjugating/ligating enzymatic activity and the specific cysteines involved. We then discuss the consequences when this activity is partially inhibited in sickle cell disease and the possibility that the same attenuation is occurring in multiple organ dysfunction syndrome. We finish by discussing the reasons for believing that nonerythroid α spectrin isoforms (I and II) also have this activity and the importance of testing this hypothesis. If correct, this would suggest that the nonerythroid spectrin isoforms play a major role in protein ubiquitination in all cell types. This would open new fields in experimental biology focused on uncovering the impact that this enzymatic activity has upon protein-protein interactions, protein turnover, cellular signaling, and many other functions impacted by spectrin, including DNA repair.
Collapse
Affiliation(s)
- Steven R Goodman
- Department of Biochemistry and Molecular Biology, Department of Pediatrics, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Rachel Petrofes Chapa
- Department of Medical Physiology, College of Medicine, Texas A&M Health Science Center, College Station, TX 77843, USA
| | - Warren E Zimmer
- Department of Medical Physiology, College of Medicine, Texas A&M Health Science Center, College Station, TX 77843, USA
| |
Collapse
|
13
|
Sukriti S, Tauseef M, Yazbeck P, Mehta D. Mechanisms regulating endothelial permeability. Pulm Circ 2015; 4:535-51. [PMID: 25610592 DOI: 10.1086/677356] [Citation(s) in RCA: 209] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 03/03/2014] [Indexed: 12/26/2022] Open
Abstract
The endothelial monolayer partitioning underlying tissue from blood components in the vessel wall maintains tissue fluid balance and host defense through dynamically opening intercellular junctions. Edemagenic agonists disrupt endothelial barrier function by signaling the opening of the intercellular junctions leading to the formation of protein-rich edema in the interstitial tissue, a hallmark of tissue inflammation that, if left untreated, causes fatal diseases, such as acute respiratory distress syndrome. In this review, we discuss how intercellular junctions are maintained under normal conditions and after stimulation of endothelium with edemagenic agonists. We have focused on reviewing the new concepts dealing with the alteration of adherens junctions after inflammatory stimulus.
Collapse
Affiliation(s)
- Sukriti Sukriti
- Department of Pharmacology and Center for Lung and Vascular Biology, College of Medicine, University of Illinois, Chicago, Illinois, USA
| | - Mohammad Tauseef
- Department of Pharmacology and Center for Lung and Vascular Biology, College of Medicine, University of Illinois, Chicago, Illinois, USA
| | - Pascal Yazbeck
- Department of Pharmacology and Center for Lung and Vascular Biology, College of Medicine, University of Illinois, Chicago, Illinois, USA
| | - Dolly Mehta
- Department of Pharmacology and Center for Lung and Vascular Biology, College of Medicine, University of Illinois, Chicago, Illinois, USA
| |
Collapse
|
14
|
Xu N, Cioffi DL, Alexeyev M, Rich TC, Stevens T. Sodium entry through endothelial store-operated calcium entry channels: regulation by Orai1. Am J Physiol Cell Physiol 2014; 308:C277-88. [PMID: 25428882 DOI: 10.1152/ajpcell.00063.2014] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Orai1 interacts with transient receptor potential protein of the canonical subfamily (TRPC4) and contributes to calcium selectivity of the endothelial cell store-operated calcium entry current (ISOC). Orai1 silencing increases sodium permeability and decreases membrane-associated calcium, although it is not known whether Orai1 is an important determinant of cytosolic sodium transitions. We test the hypothesis that, upon activation of store-operated calcium entry channels, Orai1 is a critical determinant of cytosolic sodium transitions. Activation of store-operated calcium entry channels transiently increased cytosolic calcium and sodium, characteristic of release from an intracellular store. The sodium response occurred more abruptly and returned to baseline more rapidly than did the transient calcium rise. Extracellular choline substitution for sodium did not inhibit the response, although 2-aminoethoxydiphenyl borate and YM-58483 reduced it by ∼50%. After this transient response, cytosolic sodium continued to increase due to influx through activated store-operated calcium entry channels. The magnitude of this sustained increase in cytosolic sodium was greater when experiments were conducted in low extracellular calcium and when Orai1 expression was silenced; these two interventions were not additive, suggesting a common mechanism. 2-Aminoethoxydiphenyl borate and YM-58483 inhibited the sustained increase in cytosolic sodium, only in the presence of Orai1. These studies demonstrate that sodium permeates activated store-operated calcium entry channels, resulting in an increase in cytosolic sodium; the magnitude of this response is determined by Orai1.
Collapse
Affiliation(s)
- Ningyong Xu
- Department of Pharmacology, University of South Alabama, Mobile, Alabama; Center for Lung Biology, University of South Alabama, Mobile, Alabama
| | - Donna L Cioffi
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, Alabama; Center for Lung Biology, University of South Alabama, Mobile, Alabama
| | - Mikhail Alexeyev
- Department of Cell Biology and Neuroscience, University of South Alabama, Mobile, Alabama; Center for Lung Biology, University of South Alabama, Mobile, Alabama
| | - Thomas C Rich
- Department of Pharmacology, University of South Alabama, Mobile, Alabama; Center for Lung Biology, University of South Alabama, Mobile, Alabama
| | - Troy Stevens
- Department of Pharmacology, University of South Alabama, Mobile, Alabama; Department of Medicine, University of South Alabama, Mobile, Alabama; and Center for Lung Biology, University of South Alabama, Mobile, Alabama
| |
Collapse
|
15
|
Vasauskas AA, Chen H, Wu S, Cioffi DL. The serine-threonine phosphatase calcineurin is a regulator of endothelial store-operated calcium entry. Pulm Circ 2014; 4:116-27. [PMID: 25006427 DOI: 10.1086/675641] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Accepted: 01/15/2014] [Indexed: 01/25/2023] Open
Abstract
Disruption of the endothelium leads to increased permeability, allowing extravasation of macromolecules and other solutes from blood vessels. Calcium entry through a calcium-selective, store-operated calcium (SOC) channel, I soc, contributes to barrier disruption. An understanding of the mechanisms surrounding the regulation of I soc is far from complete. We show that the calcium/calmodulin-activated phosphatase calcineurin (CN) plays a role in regulation of SOC entry, possibly through the dephosphorylation of stromal interaction molecule 1 (STIM1). Phosphorylation has been implicated as a regulatory mechanism of activity for a number of canonical transient receptor potential (TRPC) and SOC channels, including I soc. Our results show that STIM1 phosphorylation increases in pulmonary artery endothelial cells (PAECs) upon activation of SOC entry. However, the phosphatases involved in STIM1 dephosphorylation are unknown. We found that a CN inhibitor (calcineurin inhibitory peptide [CIP]) increases the phosphorylation pattern of STIM1. Using a fura 2-acetoxymethyl ester approach to measure cytosolic calcium in PAECs, we found that CIP decreases SOC entry following thapsigargin treatment in PAECs. Luciferase assays indicate that thapsigargin induces activation of CN activity and confirm inhibition of CN activity by CIP in PAECs. Also, I soc is significantly attenuated in whole-cell patch-clamp studies of PAECs treated with CIP. Finally, PAECs pretreated with CIP exhibit decreased interendothelial cell gap formation in response to thapsigargin-induced SOC entry, as compared to control cells. Taken together, our data show that CN contributes to the phosphorylation status of STIM1, which is important in regulation of endothelial SOC entry and I soc activity.
Collapse
Affiliation(s)
- Audrey A Vasauskas
- Departments of Biochemistry and Molecular Biology and Center for Lung Biology, University of South Alabama, Mobile, Alabama, USA
| | - Hairu Chen
- Department of Anesthesiology and Perioperative Medicine, Georgia Regents University, Augusta, Georgia, USA
| | - Songwei Wu
- Department of Anesthesiology and Perioperative Medicine, Georgia Regents University, Augusta, Georgia, USA
| | - Donna L Cioffi
- Departments of Biochemistry and Molecular Biology and Center for Lung Biology, University of South Alabama, Mobile, Alabama, USA
| |
Collapse
|
16
|
O'Brien ET, Wang Y, Ying H, Yue BYJT. Differential expression of genes in cells cultured from juxtacanalicular trabecular meshwork and Schlemm's canal. J Ocul Pharmacol Ther 2014; 30:291-9. [PMID: 24611521 DOI: 10.1089/jop.2013.0189] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
PURPOSE The purpose of this study was to distinguish differences in gene expression between cells cultured from the juxtacanalicular trabecular meshwork (JCTM) and those from Schlemm's canal (SC), to gain clues to differences between those cell types, and to add to our baseline knowledge of gene expression differences in these cell types for later comparison between cells from nonprimary open-angle glaucoma (POAG) and POAG outflow tissues. METHODS A set of JCTM and SC cells was cultured from each of 2 donor eyes by an explant method, grown to passage 3, and frozen in liquid nitrogen. The cells were thawed, total RNA was extracted, and the probes made from total RNAs were hybridized to MICROMAX human cDNA microarray slides in 2 separate trials. Differentially expressed genes were analyzed using PubMed, Prosite, and IPA software, and the expression of several of the genes including intercellular adhesion molecule-1 (ICAM-1), tenascin, and β-spectrin was assessed by immunofluorescence. RESULTS Schlemm's canal cells differentially expressed ICAM-1, spectrin, complement, fibulin-1, and several genes consistent with an endothelial origin in both arrays, while the JCTM cells more often overexpressed genes consistent with contractile, matrix function, and neural character. At the same time, many genes highly expressed in the first array were not highly overexpressed in the second. One highly overexpressed gene in the JCTM in both arrays, that for heparan sulfate 3-O-sulfotransferase-1 precursor, is thought to be somewhat unique, and could affect the glycosaminoglycan functionality in the extracellular matrix (ECM). CONCLUSIONS We found generally good agreement between the 2 array trials, but some contradictions as well. Many of the genes overexpressed in each cell type had been described in earlier work, but several were new. Tables of genes, grouped by cellular function, and the complete datasets are provided for the development of new hypotheses.
Collapse
Affiliation(s)
- E Timothy O'Brien
- 1 Department of Physics and Astronomy, University of North Carolina , Chapel Hill, North Carolina
| | | | | | | |
Collapse
|
17
|
Zhang R, Zhang C, Zhao Q, Li D. Spectrin: structure, function and disease. SCIENCE CHINA-LIFE SCIENCES 2013; 56:1076-85. [PMID: 24302288 DOI: 10.1007/s11427-013-4575-0] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2012] [Accepted: 03/20/2013] [Indexed: 01/23/2023]
Abstract
Spectrin is a large, cytoskeletal, and heterodimeric protein composed of modular structure of α and β subunits, it typically contains 106 contiguous amino acid sequence motifs called "spectrin repeats". Spectrin is crucial for maintaining the stability and structure of the cell membrane and the shape of a cell. Moreover, it contributes to diverse cell functions such as cell adhesion, cell spreading, and the cell cycle. Mutations of spectrin lead to various human diseases such as hereditary hemolytic anemia, type 5 spinocerebellar ataxia, cancer, as well as others. This review focuses on recent advances in determining the structure and function of spectrin as well as its role in disease.
Collapse
Affiliation(s)
- Rui Zhang
- Jiangsu Engineering Research Center for microRNA Biology and Biotechnology, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210093, China
| | | | | | | |
Collapse
|
18
|
Kadeba PI, Vasauskas AA, Chen H, Wu S, Scammell JG, Cioffi DL. Regulation of store-operated calcium entry by FK506-binding immunophilins. Cell Calcium 2013; 53:275-85. [PMID: 23375350 DOI: 10.1016/j.ceca.2012.12.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2012] [Revised: 12/21/2012] [Accepted: 12/24/2012] [Indexed: 02/06/2023]
Abstract
Calcium entry from the extracellular space into cells is an important signaling mechanism in both physiological and pathophysiological functions. In non-excitable cells, store-operated calcium (SOC) entry represents a principal mode of calcium entry. Activation of SOC entry in pulmonary artery endothelial cells leads to the formation of inter-endothelial cell gaps and subsequent endothelial barrier disruption. Regulation of endothelial SOC entry is poorly understood. In this work, we identify two large molecular weight immunophilins, FKBP51 and FKBP52, as novel regulators of SOC entry in endothelial cells. Using cell fractionation studies and immunocytochemistry we determined that a fraction of these largely cytosolic proteins localize to the plasma membrane where SOC entry channels are found. That FKBP51 and FKBP52 associate with SOC entry channel protein complexes was supported by co-precipitation of the immunophilins with TRPC4, a subunit of the calcium-selective, SOC entry channel ISOC. Dexamethasone-induced upregulation of FKBP51 expression in pulmonary artery endothelial cells reduced global SOC entry as well as ISOC. Similar results were observed when FKBP51 was over-expressed in an inducible HEK293 cell line. On the other hand, when FKBP52 was over-expressed SOC entry was enhanced. When expression of FKBP52 was inhibited, SOC entry was decreased. Collectively, our observations support regulatory roles for these large molecular weight immunophilins in which FKBP51 inhibits, whereas FKBP52 enhances, SOC entry in endothelial cells.
Collapse
Affiliation(s)
- Pierre I Kadeba
- Department of Biochemistry, University of South Alabama, Mobile, AL 36688, United States
| | | | | | | | | | | |
Collapse
|
19
|
Vanoverberghe K, Lehen’kyi V, Thébault S, Raphaël M, Vanden Abeele F, Slomianny C, Mariot P, Prevarskaya N. Cytoskeleton reorganization as an alternative mechanism of store-operated calcium entry control in neuroendocrine-differentiated cells. PLoS One 2012; 7:e45615. [PMID: 23049826 PMCID: PMC3458093 DOI: 10.1371/journal.pone.0045615] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2012] [Accepted: 08/23/2012] [Indexed: 02/07/2023] Open
Abstract
Neuroendocrine differentiation (NED) is a hallmark of advanced androgen-independent prostate cancer, for which no successful therapy exists. NED tumour cells escape apoptotic cell death by alterations of Ca(2+) homeostasis where the store-operated Ca(2+) entry (SOCE) is known to be a key event. We have previously shown that the downregulation of Orai1 protein representing the major molecular component of endogenous SOCE in human prostate cancer cells, and constituting the principal source of Ca(2+) influx used by the cell to trigger apoptosis, contributes to the establishment of an apoptosis-resistant phenotype (Cell Death Dis. 2010 Sep 16;1:e75.). Here, we report for the first time that the decrease of SOCE during NED may be caused by alternative NED-induced mechanism involving cytoskeleton reorganisation. NED induced by androgen deprivation resulted in a decrease of SOCE due to cortical F-actin over-polymerization which inhibits thapsigargin-induced SOCE. The disruption of F-actin polymerization by Cytochalasin D in NED cells restored SOCE, while the induction of F-actin polymerization by jasplakinolide or calyculin A diminished SOCE without changing the expression of key SOCE players: Orai1, STIM1, and TRPC1. Our data suggest that targeting cytoskeleton-induced pathways of malignant cells together with SOCE-involved channels may prove a useful strategy in the treatment of advanced prostate cancer.
Collapse
Affiliation(s)
- Karine Vanoverberghe
- Inserm, U-1003, Equipe labellisée par la Ligue Nationale contre le cancer, Villeneuve d’Ascq, France
- Laboratory of Excellence, Ion Channels Science and Therapeutics, Université des Sciences et Technologies de Lille (USTL), Villeneuve d’Ascq, France
| | - V’yacheslav Lehen’kyi
- Inserm, U-1003, Equipe labellisée par la Ligue Nationale contre le cancer, Villeneuve d’Ascq, France
- Laboratory of Excellence, Ion Channels Science and Therapeutics, Université des Sciences et Technologies de Lille (USTL), Villeneuve d’Ascq, France
| | - Stéphanie Thébault
- Inserm, U-1003, Equipe labellisée par la Ligue Nationale contre le cancer, Villeneuve d’Ascq, France
- Laboratory of Excellence, Ion Channels Science and Therapeutics, Université des Sciences et Technologies de Lille (USTL), Villeneuve d’Ascq, France
| | - Maylis Raphaël
- Inserm, U-1003, Equipe labellisée par la Ligue Nationale contre le cancer, Villeneuve d’Ascq, France
- Laboratory of Excellence, Ion Channels Science and Therapeutics, Université des Sciences et Technologies de Lille (USTL), Villeneuve d’Ascq, France
| | - Fabien Vanden Abeele
- Inserm, U-1003, Equipe labellisée par la Ligue Nationale contre le cancer, Villeneuve d’Ascq, France
- Laboratory of Excellence, Ion Channels Science and Therapeutics, Université des Sciences et Technologies de Lille (USTL), Villeneuve d’Ascq, France
| | - Christian Slomianny
- Inserm, U-1003, Equipe labellisée par la Ligue Nationale contre le cancer, Villeneuve d’Ascq, France
- Laboratory of Excellence, Ion Channels Science and Therapeutics, Université des Sciences et Technologies de Lille (USTL), Villeneuve d’Ascq, France
| | - Pascal Mariot
- Inserm, U-1003, Equipe labellisée par la Ligue Nationale contre le cancer, Villeneuve d’Ascq, France
- Laboratory of Excellence, Ion Channels Science and Therapeutics, Université des Sciences et Technologies de Lille (USTL), Villeneuve d’Ascq, France
| | - Natalia Prevarskaya
- Inserm, U-1003, Equipe labellisée par la Ligue Nationale contre le cancer, Villeneuve d’Ascq, France
- Laboratory of Excellence, Ion Channels Science and Therapeutics, Université des Sciences et Technologies de Lille (USTL), Villeneuve d’Ascq, France
- * E-mail:
| |
Collapse
|
20
|
Kim H, Kim J, Jeon JP, Myeong J, Wie J, Hong C, Kim HJ, Jeon JH, So I. The roles of G proteins in the activation of TRPC4 and TRPC5 transient receptor potential channels. Channels (Austin) 2012; 6:333-43. [PMID: 22878724 PMCID: PMC3508772 DOI: 10.4161/chan.21198] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
TRPC4 and TRPC5 channels are important regulators of electrical excitability in both gastrointestinal myocytes and neurons. Much is known regarding the assembly and function of these channels including TRPC1 as a homotetramer or a heteromultimer and the roles that their interacting proteins play in controlling these events. Further, they are one of the best-studied targets of G protein-coupled receptors and growth factors in general and Gαq protein coupled receptor or epidermal growth factor in particular. However, our understanding of the roles of Gαi/o proteins on TRPC4/5 channels is still rudimentary. We discuss potential roles for Gαi/o proteins in channel activation in addition to their known role in cellular signaling.
Collapse
Affiliation(s)
- Hana Kim
- Department of Physiology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Cioffi DL, Wu S, Chen H, Alexeyev M, St Croix CM, Pitt BR, Uhlig S, Stevens T. Orai1 determines calcium selectivity of an endogenous TRPC heterotetramer channel. Circ Res 2012; 110:1435-44. [PMID: 22534489 DOI: 10.1161/circresaha.112.269506] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE Canonical transient receptor potential 4 (TRPC4) contributes to the molecular composition of a channel encoding for a calcium selective store-operated current, I(SOC), whereas Orai1 critically comprises a channel encoding for the highly selective calcium release activated calcium current, I(CRAC). However, Orai1 may interact with TRPC proteins and influence their activation and permeation characteristics. Endothelium expresses both TRPC4 and Orai1, and it remains unclear as to whether Orai1 interacts with TRPC4 and contributes to calcium permeation through the TPRC4 channel. OBJECTIVE We tested the hypothesis that Orai1 interacts with TRPC4 and contributes to the channel's selective calcium permeation important for endothelial barrier function. METHODS AND RESULTS A novel method to purify the endogenous TRPC4 channel and probe for functional interactions was developed, using TRPC4 binding to protein 4.1 as bait. Isolated channel complexes were conjugated to anti-TRPC protein antibodies labeled with cy3-cy5 pairs. Förster Resonance Energy Transfer among labeled subunits revealed the endogenous protein alignment. One TRPC1 and at least 2 TRPC4 subunits constituted the endogenous channel (TRPC1/4). Orai1 interacted with TRPC4. Conditional Orai1 knockdown reduced the probability for TRPC1/4 channel activation and converted it from a calcium-selective to a nonselective channel, an effect that was rescued on Orai1 reexpression. Loss of Orai1 improved endothelial cell barrier function. CONCLUSION Orai1 interacts with TRPC4 in the endogenous channel complex, where it controls TRPC1/4 activation and channel permeation characteristics, including calcium selectivity, important for control of endothelial cell barrier function.
Collapse
Affiliation(s)
- Donna L Cioffi
- Department of Biochemistry and Molecular Biology, University of South Alabama College of Medicine, Mobile, AL 36688, USA. dlcioffi@ usouthal.edu
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Machnicka B, Grochowalska R, Bogusławska DM, Sikorski AF, Lecomte MC. Spectrin-based skeleton as an actor in cell signaling. Cell Mol Life Sci 2011; 69:191-201. [PMID: 21877118 PMCID: PMC3249148 DOI: 10.1007/s00018-011-0804-5] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2011] [Revised: 08/08/2011] [Accepted: 08/10/2011] [Indexed: 01/12/2023]
Abstract
This review focuses on the recent advances in functions of spectrins in non-erythroid cells. We discuss new data concerning the commonly known role of the spectrin-based skeleton in control of membrane organization, stability and shape, and tethering protein mosaics to the cellular motors and to all major filament systems. Particular effort has been undertaken to highlight recent advances linking spectrin to cell signaling phenomena and its participation in signal transduction pathways in many cell types.
Collapse
Affiliation(s)
- B Machnicka
- University of Zielona Góra, Zielona Góra, Poland
| | | | | | | | | |
Collapse
|
23
|
Store-operated calcium entry channels in pulmonary endothelium: the emerging story of TRPCS and Orai1. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 661:137-54. [PMID: 20204728 DOI: 10.1007/978-1-60761-500-2_9] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Cells of diverse origin utilize shifts in cytosolic calcium concentrations as intracellular signals to elicit physiological responses. In endothelium, inflammatory first messengers increase cytosolic calcium as a signal to disrupt cell-cell borders and produce inter-cellular gaps. Calcium influx across the plasma membrane is required to initiate barrier disruption, although the calcium entry mechanism responsible for this effect remains poorly understood. This chapter highlights recent efforts to define the molecular anatomy of the ion channel responsible for triggering endothelial cell gap formation. Resolving the identity and function of this calcium channel will pave the way for new anti-inflammatory therapeutic targets.
Collapse
|
24
|
Cioffi DL, Lowe K, Alvarez DF, Barry C, Stevens T. TRPing on the lung endothelium: calcium channels that regulate barrier function. Antioxid Redox Signal 2009; 11:765-76. [PMID: 18783312 PMCID: PMC2850299 DOI: 10.1089/ars.2008.2221] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Rises in cytosolic calcium are sufficient to initiate the retraction of endothelial cell borders and to increase macromolecular permeability. Although endothelial cell biologists have recognized the importance of shifts in cytosolic calcium for several decades, only recently have we gained a rudimentary understanding of the membrane calcium channels that change cell shape. Members of the transient receptor potential family (TRP) are chief among the molecular candidates for permeability-coupled calcium channels. Activation of calcium entry through store-operated calcium entry channels, most notably TRPC1 and TRPC4, increases lung endothelial cell permeability, as does activation of calcium entry through the TRPV4 channel. However, TRPC1 and TRPC4 channels appear to influence the lung extraalveolar endothelial barrier most prominently, whereas TRPV4 channels appear to influence the lung capillary endothelial barrier most prominently. Thus, phenotypic heterogeneity in ion channel expression and function exists within the lung endothelium, along the arterial-capillary-venous axis, and is coupled to discrete control of endothelial barrier function.
Collapse
Affiliation(s)
- Donna L Cioffi
- Center for Lung Biology, University of South Alabama, Mobile, Alabama 36688, USA
| | | | | | | | | |
Collapse
|
25
|
Abramowitz J, Birnbaumer L. Physiology and pathophysiology of canonical transient receptor potential channels. FASEB J 2009; 23:297-328. [PMID: 18940894 PMCID: PMC2630793 DOI: 10.1096/fj.08-119495] [Citation(s) in RCA: 244] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2008] [Accepted: 09/25/2008] [Indexed: 11/11/2022]
Abstract
The existence of a mammalian family of TRPC ion channels, direct homologues of TRP, the visual transduction channel of flies, was discovered during 1995-1996 as a consequence of research into the mechanism by which the stimulation of the receptor-Gq-phospholipase Cbeta signaling pathway leads to sustained increases in intracellular calcium. Mammalian TRPs, TRPCs, turned out to be nonselective, calcium-permeable cation channels, which cause both a collapse of the cell's membrane potential and entry of calcium. The family comprises 7 members and is widely expressed. Many cells and tissues express between 3 and 4 of the 7 TRPCs. Despite their recent discovery, a wealth of information has accumulated, showing that TRPCs have widespread roles in almost all cells studied, including cells from excitable and nonexcitable tissues, such as the nervous and cardiovascular systems, the kidney and the liver, and cells from endothelia, epithelia, and the bone marrow compartment. Disruption of TRPC function is at the root of some familial diseases. More often, TRPCs are contributing risk factors in complex diseases. The present article reviews what has been uncovered about physiological roles of mammalian TRPC channels since the time of their discovery. This analysis reveals TRPCs as major and unsuspected gates of Ca(2+) entry that contribute, depending on context, to activation of transcription factors, apoptosis, vascular contractility, platelet activation, and cardiac hypertrophy, as well as to normal and abnormal cell proliferation. TRPCs emerge as targets for a thus far nonexistent field of pharmacological intervention that may ameliorate complex diseases.
Collapse
Affiliation(s)
- Joel Abramowitz
- Transmembrane Signaling Group, Laboratory of Neurobiology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA.
| | | |
Collapse
|
26
|
Prasain N, Stevens T. The actin cytoskeleton in endothelial cell phenotypes. Microvasc Res 2008; 77:53-63. [PMID: 19028505 DOI: 10.1016/j.mvr.2008.09.012] [Citation(s) in RCA: 211] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2008] [Accepted: 09/26/2008] [Indexed: 10/21/2022]
Abstract
Endothelium forms a semi-permeable barrier that separates blood from the underlying tissue. Barrier function is largely determined by cell-cell and cell-matrix adhesions that define the limits of cell borders. Yet, such cell-cell and cell-matrix tethering is critically reliant upon the nature of adherence within the cell itself. Indeed, the actin cytoskeleton fulfills this essential function, to provide a strong, dynamic intracellular scaffold that organizes integral membrane proteins with the cell's interior, and responds to environmental cues to orchestrate appropriate cell shape. The actin cytoskeleton is comprised of three distinct, but inter-related structures, including actin cross-linking of spectrin within the membrane skeleton, the cortical actin rim, and actomyosin-based stress fibers. This review addresses each of these actin-based structures, and discusses cellular signals that control the disposition of actin in different endothelial cell phenotypes.
Collapse
Affiliation(s)
- Nutan Prasain
- Department of Molecular and Cellular Pharmacology, College of Medicine, University of South Alabama, Mobile, AL 36688, USA
| | | |
Collapse
|
27
|
Yin J, Hoffmann J, Kaestle SM, Neye N, Wang L, Baeurle J, Liedtke W, Wu S, Kuppe H, Pries AR, Kuebler WM. Negative-feedback loop attenuates hydrostatic lung edema via a cGMP-dependent regulation of transient receptor potential vanilloid 4. Circ Res 2008; 102:966-74. [PMID: 18323527 DOI: 10.1161/circresaha.107.168724] [Citation(s) in RCA: 105] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Although the formation of hydrostatic lung edema is generally attributed to imbalanced Starling forces, recent data show that lung endothelial cells respond to increased vascular pressure and may thus regulate vascular permeability and edema formation. In combining real-time optical imaging of the endothelial Ca(2+) concentration ([Ca(2+)](i)) and NO production with filtration coefficient (K(f)) measurements in the isolated perfused lung, we identified a series of endothelial responses that constitute a negative-feedback loop to protect the microvascular barrier. Elevation of lung microvascular pressure was shown to increase endothelial [Ca(2+)](i) via activation of transient receptor potential vanilloid 4 (TRPV4) channels. The endothelial [Ca(2+)](i) transient increased K(f) via activation of myosin light-chain kinase and simultaneously stimulated NO synthesis. In TRPV4 deficient mice, pressure-induced increases in endothelial [Ca(2+)](i), NO synthesis, and lung wet/dry weight ratio were largely blocked. Endothelial NO formation limited the permeability increase by a cGMP-dependent attenuation of the pressure-induced [Ca(2+)](i) response. Inactivation of TRPV4 channels by cGMP was confirmed by whole-cell patch-clamp of pulmonary microvascular endothelial cells and intravital imaging of endothelial [Ca(2+)](i). Hence, pressure-induced endothelial Ca(2+) influx via TRPV4 channels increases lung vascular permeability yet concomitantly activates an NO-mediated negative-feedback loop that protects the vascular barrier by a cGMP-dependent attenuation of the endothelial [Ca(2+)](i) response. The identification of this novel regulatory pathway gives rise to new treatment strategies, as demonstrated in vivo in rats with acute myocardial infarction in which inhibition of cGMP degradation by the phosphodiesterase 5 inhibitor sildenafil reduced hydrostatic lung edema.
Collapse
Affiliation(s)
- Jun Yin
- Institute of Physiology, Charité-Universitaetsmedizin Berlin, Campus Benjamin Franklin, Arnimallee 22, 14195 Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Odell AF, Van Helden DF, Scott JL. The spectrin cytoskeleton influences the surface expression and activation of human transient receptor potential channel 4 channels. J Biol Chem 2007; 283:4395-407. [PMID: 18048348 DOI: 10.1074/jbc.m709729200] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Despite over a decade of research, only recently have the mechanisms governing transient receptor potential channel (TRPC) channel function begun to emerge, with an essential role for accessory proteins in this process. We previously identified a tyrosine phosphorylation event as critical in the plasma membrane translocation and activation of hTRPC4 channels following epidermal growth factor (EGF) receptor activation. To further characterize the signaling events underlying this process, a yeast-two hybrid screen was performed on the C terminus of hTRPC4. The intracellular C-terminal region from proline 686 to leucine 977 was used to screen a human brain cDNA library. Two members of the spectrin family, alphaII- and betaV-spectrin, were identified as binding partners. The interaction of hTRPC4 with alphaII-spectrin and betaV-spectrin was confirmed by glutathione S-transferase pulldown and co-immunoprecipitation experiments. Deletion analysis identified amino acids 730-758 of hTRPC4 as critical for the interaction with this region located within a coiled-coil domain, juxtaposing the Ca(2+)/calmodulin- and IP(3)R-binding region (CIRB domain). This region is deleted in the proposed deltahTRPC4 splice variant form, which failed to undergo both EGF-induced membrane insertion and activation, providing a genetic mechanism for regulating channel activity. We also demonstrate that the exocytotic insertion and activation of hTRPC4 following EGF application is accompanied by dissociation from alphaII-spectrin. Furthermore, depletion of alphaII-spectrin by small interference RNA reduces the basal surface expression of alphahTRPC4 and prevents the enhanced membrane insertion in response to EGF application. Importantly, depletion of alphaII-spectrin did not affect the expression of the delta variant. Taken together, these results demonstrate that a direct interaction between hTRPC4 and the spectrin cytoskeleton is involved in the regulation of hTRPC4 surface expression and activation.
Collapse
Affiliation(s)
- Adam F Odell
- School of Biomedical Sciences, Faculty of Health, University of Newcastle, Level 5, MSB, University Drive, New South Wales 2308, Australia.
| | | | | |
Collapse
|
29
|
Wang J, Weigand L, Foxson J, Shimoda LA, Sylvester JT. Ca2+ signaling in hypoxic pulmonary vasoconstriction: effects of myosin light chain and Rho kinase antagonists. Am J Physiol Lung Cell Mol Physiol 2007; 293:L674-85. [PMID: 17575009 DOI: 10.1152/ajplung.00141.2007] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Antagonists of myosin light chain (MLC) kinase (MLCK) and Rho kinase (ROK) are thought to inhibit hypoxic pulmonary vasoconstriction (HPV) by decreasing the concentration of phosphorylated MLC at any intracellular Ca(2+) concentration ([Ca(2+)](i)) in pulmonary arterial smooth muscle cells (PASMC); however, these antagonists can also decrease [Ca(2+)](i). To determine whether MLCK and ROK antagonists alter Ca(2+) signaling in HPV, we measured the effects of ML-9, ML-7, Y-27632, and HA-1077 on [Ca(2+)](i), Ca(2+) entry, and Ca(2+) release in rat distal PASMC exposed to hypoxia or depolarizing concentrations of KCl. We performed parallel experiments in isolated rat lungs to confirm the inhibitory effects of these agents on pulmonary vasoconstriction. Our results demonstrate that MLCK and ROK antagonists caused concentration-dependent inhibition of hypoxia-induced increases in [Ca(2+)](i) in PASMC and HPV in isolated lungs and suggest that this inhibition was due to blockade of Ca(2+) release from the sarcoplasmic reticulum and Ca(2+) entry through store- and voltage-operated Ca(2+) channels in PASMC. Thus MLCK and ROK antagonists might block HPV by inhibiting Ca(2+) signaling, as well as the actin-myosin interaction, in PASMC. If effects on Ca(2+) signaling were due to decreased phosphorylated myosin light chain concentration, their diversity suggests that MLCK and ROK antagonists may have acted by inhibiting myosin motors and/or altering the cytoskeleton in a manner that prevented achievement of required spatial relationships among the cellular components of the response.
Collapse
Affiliation(s)
- Jian Wang
- Division of Pulmonary & Critical Care Medicine, The Johns Hopkins Asthma and Allergy Center, Baltimore, MD 21224, USA
| | | | | | | | | |
Collapse
|
30
|
Abstract
In recent decades, it has become evident that the endothelium is by no means a passive inner lining of blood vessels. This 'organ' with a large surface (approximately 350 m2) and a comparatively small total mass (approximately 110 g) is actively involved in vital functions of the cardiovascular system, including regulation of perfusion, fluid and solute exchange, haemostasis and coagulation, inflammatory responses, vasculogenesis and angiogenesis. The present chapter focusses on two central aspects of endothelial structure and function: (1) the heterogeneity in endothelial properties between species, organs, vessel classes and even within individual vessels and (2) the composition and role of the molecular layer on the luminal surface of endothelial cells. The endothelial lining of blood vessels in different organs differs with respect to morphology and permeability and is classified as 'continuous', 'fenestrated' or 'discontinuous'. Furthermore, the mediator release, antigen presentation or stress responses of endothelial cells vary between species, different organs and vessel classes. Finally there are relevant differences even between adjacent endothelial cells, with some cells exhibiting specific functional properties, e.g. as pacemaker cells for intercellular calcium signals. Organ-specific structural and functional properties of the endothelium are marked in the vascular beds of the lung and the brain. Pulmonary endothelium exhibits a high constitutive expression of adhesion molecules which may contribute to the margination of the large intravascular pool of leucocytes in the lung. Furthermore, the pulmonary microcirculation is less permeable to protein and water flux as compared to large pulmonary vessels. Endothelial cells of the blood-brain barrier exhibit a specialised phenotype with no fenestrations, extensive tight junctions and sparse pinocytotic vesicular transport. This barrier allows a strict control of exchange of solutes and circulating cells between the plasma and the interstitial space. It was observed that average haematocrit levels in muscle capillaries are much lower as compared to systemic haematocrit, and that flow resistance of microvascular beds is higher than expected from in vitro studies of blood rheology. This evidence stimulated the concept of a substantial layer on the luminal endothelial surface (endothelial surface layer, ESL) with a thickness in the range of 0.5-1 microm. In comparison, the typical thickness of the glycocalyx directly anchored in the endothelial plasma membrane, as seen in electron micrographs, amounts to only about 50-100 microm. Therefore it is assumed that additional components, e.g. adsorbed plasma proteins or hyaluronan, are essential in constituting the ESL. Functional consequences of the ESL presence are not yet sufficiently understood and acknowledged. However, it is evident that the thick endothelial surface layer significantly impacts haemodynamic conditions, mechanical stresses acting on red cells in microvessels, oxygen transport, vascular control, coagulation, inflammation and atherosclerosis.
Collapse
Affiliation(s)
- A R Pries
- Dept. of Physiology, Charité Berlin, Arnimallee 22, 14195 Berlin, Germany.
| | | |
Collapse
|
31
|
Abstract
Elevations in cytosolic Ca2+ concentration are the usual initial response of endothelial cells to hormonal and chemical transmitters and to changes in physical parameters, and many endothelial functions are dependent upon changes in Ca2+ signals produced. Endothelial cell Ca2+ signalling shares similar features with other electrically non-excitable cell types, but has features unique to endothelial cells. This chapter discusses the major components of endothelial cell Ca2+ signalling.
Collapse
Affiliation(s)
- Q K Tran
- Division of Molecular Biology and Biochemistry, School of Biological Sciences, University of Missouri-Kansas City, 5007 Rockhill Road, Kansas City, MO 64110, USA
| | | |
Collapse
|
32
|
Zhou C, Chen H, Lu F, Sellak H, Daigle JA, Alexeyev MF, Xi Y, Ju J, van Mourik JA, Wu S. Cav3.1 (alpha1G) controls von Willebrand factor secretion in rat pulmonary microvascular endothelial cells. Am J Physiol Lung Cell Mol Physiol 2006; 292:L833-44. [PMID: 17172292 PMCID: PMC3048959 DOI: 10.1152/ajplung.00377.2006] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
The T-type Ca2+ channel Cav3.1 subunit is present in pulmonary microvascular endothelial cells (PMVECs), but not in pulmonary artery endothelial cells (PAECs). The present study sought to assess the role of Cav3.1 in thrombin-induced Weibel-Palade body exocytosis and consequent von Willebrand factor (VWF) release. In PMVECs and PAECs transduced with a green fluorescent protein (GFP)-tagged VWF chimera, we examined the real-time dynamics and secretory process of VWF-GFP-containing vesicles in response to thrombin and the cAMP-elevating agent isoproterenol. Whereas thrombin stimulated a progressive decrease in the number of VWF-GFP-containing vesicles in both cell types, isoproterenol only decreased the number of VWF-GFP-containing vesicles in PAECs. In PMVECs, thrombin-induced decrease in the number of VWF-GFP-containing vesicles was nearly abolished by the T-type Ca2+ channel blocker mibefradil as well as by Cav3.1 gene silencing with small hairpin RNA. Expression of recombinant Cav3.1 subunit in PAECs resulted in pronounced increase in thrombin-stimulated Ca2+ entry, which is sensitive to mibefradil. Together, these data indicate that VWF secretion from lung endothelial cells is regulated by two distinct pathways involving Ca2+ or cAMP, and support the hypothesis that activation of Cav3.1 T-type Ca2+ channels in PMVECs provides a unique cytosolic Ca2+ source important for Gq-linked agonist-induced VWF release.
Collapse
Affiliation(s)
- Chun Zhou
- Center for Lung Biology, University of South Alabama College of Medicine, Mobile, AL 36688-0002, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Parker JC, Stevens T, Randall J, Weber DS, King JA. Hydraulic conductance of pulmonary microvascular and macrovascular endothelial cell monolayers. Am J Physiol Lung Cell Mol Physiol 2006; 291:L30-7. [PMID: 16760315 DOI: 10.1152/ajplung.00317.2005] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Endothelial cells isolated from pulmonary arteries (RPAEC) and microcirculation (RPMVEC) of rat lungs were grown to confluence on porous filters and mounted on an Ussing-type chamber. Transmembrane pressure (deltaP) was controlled by the reservoir height, and the filtration rate corrected for surface area (J(v)/A) was measured by timing fluid movement in a calibrated micropipette. These parameters were used to calculate hydraulic conductance (Lp) by using linear regression of J(v)/A on deltaP. Mean Lp values for newly confluent RPAEC monolayers were 22 times higher than those for RPMVEC monolayers (28.6 +/- 5.6 vs. 1.30 +/- 0.50 x 10(-7) cm x s(-1) x cmH2O(-1); P < or = 0.01). After confluence was reached, electrical resistance and Lp remained stable in RPAEC but continued to change in RPMVEC with days in culture. Both phenotypes exhibited an initial time-dependent sealing response, but Lp also had an inverse relationship to deltaP in RPMVEC monolayers > or = 4 days postconfluence that was attributed to cell overgrowth rather than junctional length. In a comparison of the cadherin contents, E-cadherin was predominant in RPMVEC, but VE-cadherin was predominant in RPAEC. At a constant deltaP of 40-45 cmH2O for 2 h, J(v)/A increased 225% in RPAEC monolayers but did not change significantly in RPMVEC monolayers. Significant decreases in Lp were obtained after treatment with 5% albumin, GdCl3, or isoproterenol plus rolipram in both phenotypes. Thus lung microvascular endothelial cells exhibited a significantly lower Lp than conduit vessel endothelium, which would limit alveolar flooding relative to perivascular edema cuff formation during increased pulmonary vascular pressures.
Collapse
Affiliation(s)
- James C Parker
- Dept. of Physiology, MSB 3074, College of Medicine, Univ. of South Alabama, Mobile, AL 36688, USA.
| | | | | | | | | |
Collapse
|
34
|
Street M, Marsh SJ, Stabach PR, Morrow JS, Brown DA, Buckley NJ. Stimulation of Gαq-coupled M1 muscarinic receptor causes reversible spectrin redistribution mediated by PLC, PKC and ROCK. J Cell Sci 2006; 119:1528-36. [PMID: 16551696 DOI: 10.1242/jcs.02872] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Spectrin is a cytoskeletal protein that plays a role in formation of the specialized plasma membrane domains. However, little is known of the molecular mechanism that regulates responses of spectrin to extracellular stimuli, such as activation of G-protein-coupled receptor (GPCR). We have found that αII spectrin is a component of the Gαq/11-associated protein complex in CHO cells stably expressing the M1 muscarinic receptor, and investigated the effect of activation of GPCR on the cellular localization of yellow-fluorescent-protein-tagged αII spectrin. Stimulation of Gαq/11-coupled M1 muscarinic receptor triggered reversible redistribution of αII spectrin following a rise in intracellular Ca2+ concentration. This redistribution, accompanied by non-apoptotic membrane blebbing, required an intact actin cytoskeleton and was dependent on activation of phospholipase C, protein kinase C, and Rho-associated kinase ROCK. Muscarinic-agonist-induced spectrin remodeling appeared particularly active at localized domains, which is clear contrast to that caused by constitutive activation of ROCK and to global rearrangement of the spectrin lattice caused by changes in osmotic pressure. These results suggest a role for spectrin in providing a dynamic and reversible signaling platform to the specific domains of the plasma membrane in response to stimulation of GPCR.
Collapse
Affiliation(s)
- Miyoko Street
- University of Leeds, Institute of Membrane and Systems Biology, Garstang Building, Mount Preston Street, Leeds LS2 9JT, UK.
| | | | | | | | | | | |
Collapse
|
35
|
Abstract
The microvascular endothelial cell monolayer localized at the critical interface between the blood and vessel wall has the vital functions of regulating tissue fluid balance and supplying the essential nutrients needed for the survival of the organism. The endothelial cell is an exquisite “sensor” that responds to diverse signals generated in the blood, subendothelium, and interacting cells. The endothelial cell is able to dynamically regulate its paracellular and transcellular pathways for transport of plasma proteins, solutes, and liquid. The semipermeable characteristic of the endothelium (which distinguishes it from the epithelium) is crucial for establishing the transendothelial protein gradient (the colloid osmotic gradient) required for tissue fluid homeostasis. Interendothelial junctions comprise a complex array of proteins in series with the extracellular matrix constituents and serve to limit the transport of albumin and other plasma proteins by the paracellular pathway. This pathway is highly regulated by the activation of specific extrinsic and intrinsic signaling pathways. Recent evidence has also highlighted the importance of the heretofore enigmatic transcellular pathway in mediating albumin transport via transcytosis. Caveolae, the vesicular carriers filled with receptor-bound and unbound free solutes, have been shown to shuttle between the vascular and extravascular spaces depositing their contents outside the cell. This review summarizes and analyzes the recent data from genetic, physiological, cellular, and morphological studies that have addressed the signaling mechanisms involved in the regulation of both the paracellular and transcellular transport pathways.
Collapse
Affiliation(s)
- Dolly Mehta
- Center of Lung and Vascular Biology, Dept. of Pharmacology (M/C 868), University of Illinois, 835 S. Wolcott Avenue, Chicago, IL 60612, USA
| | | |
Collapse
|
36
|
Tiruppathi C, Ahmmed GU, Vogel SM, Malik AB. Ca2+Signaling, TRP Channels, and Endothelial Permeability. Microcirculation 2006; 13:693-708. [PMID: 17085428 DOI: 10.1080/10739680600930347] [Citation(s) in RCA: 188] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Increased endothelial permeability is the hallmark of inflammatory vascular edema. Inflammatory mediators that bind to heptahelical G protein-coupled receptors trigger increased endothelial permeability by increasing the intracellular Ca2+ concentration ([Ca2+]i). The rise in [Ca2+]i activates key signaling pathways that mediate cytoskeletal reorganization (through myosin-light-chain-dependent contraction) and the disassembly of VE-cadherin at the adherens junctions. The Ca2+-dependent protein kinase C (PKC) isoform PKCalpha plays a crucial role in initiating endothelial cell contraction and disassembly of VE-cadherin junctions. The increase in [Ca2+]i induced by inflammatory agonists such as thrombin and histamine is achieved by the generation of inositol 1,4,5-trisphosphate (IP3), activation of IP3-receptors, release of stored intracellular Ca2+, and Ca2+ entry through plasma membrane channels. IP3-sensitive Ca2+-store depletion activates plasma membrane cation channels (i.e., store-operated cation channels [SOCs] or Ca2+ release-activated channels [CRACs]) to cause Ca2+ influx into endothelial cells. Recent studies have identified members of Drosophila transient receptor potential (TRP) gene family of channels that encode functional SOCs in endothelial cells. These studies also suggest that the canonical TRPC homologue TRPC1 is the predominant isoform expressed in human vascular endothelial cells, and is the essential component of the SOC in this cell type. Further, evidence suggests that the inflammatory cytokine tumor necrosis factor-alpha can induce the expression of TRPC1 in human vascular endothelial cells signaling via the nuclear factor-kappaB pathway. Increased expression of TRPC1 augments Ca2+ influx via SOCs and potentiates the thrombin-induced increase in permeability in human vascular endothelial cells. Deletion of the canonical TRPC homologue in mouse, TRPC4, caused impairment in store-operated Ca2+ current and Ca2+-store release-activated Ca2+ influx in aortic and lung endothelial cells. In TRPC4 knockout (TRPC4-/-) mice, acetylcholine-induced endothelium-dependent smooth muscle relaxation was drastically reduced. In addition, TRPC4-/- mouse-lung endothelial cells exhibited lack of actin-stress fiber formation and cell retraction in response to thrombin activation of protease-activated receptor-1 (PAR-1) in endothelial cells. The increase in lung microvascular permeability in response to PAR-1 activation was inhibited in TRPC4-/- mice. These results indicate that endothelial TRP channels such as TRPC1 and TRPC4 play an important role in signaling agonist-induced increases in endothelial permeability.
Collapse
Affiliation(s)
- Chinnaswamy Tiruppathi
- Department of Pharmacology and Center for Lung and Vascular Biology, College of Medicine, University of Illinois, Chicago, Illinois 60612, USA.
| | | | | | | |
Collapse
|
37
|
Sugiyama D, Arai KI, Tsuji K. Definitive Hematopoiesis from Acetyl LDL Incorporating Endothelial Cells in the Mouse Embryo. Stem Cells Dev 2005; 14:687-96. [PMID: 16433624 DOI: 10.1089/scd.2005.14.687] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Previously, we reported generation of erythropoiesis from acetyl low-density lipoprotein (Ac-LDL)- incorporating endothelial cells (ECs) in the mouse embryo. However, it is still unclear whether the other types of definitive hematopoietic cells (HCs) can be generated from these cells. In this study, ECs at 10 days post coitum (dpc) were tagged with Ac-LDL-DiI and were shown to release DiI+ HCs into the circulation after 12 h of whole embryo culture. The hematopoietic clusters in the main arteries were also stained with DiI. The circulating DiI+ HCs expressed c-Kit and half of these cells displayed blastic morphology. In vitro culture and in vivo reconstitution experiments demonstrated that the circulating DiI+ HCs contained definitive multipotent progenitors, including hematopoietic stem cells (HSCs). Furthermore, the sorted DiI+ HCs were able to colonize the fetal liver (FL) when introduced back into the blood stream of a secondary recipient embryo. These results suggest that Ac-LDL incorporating ECs can produce definitive HSCs and HCs colonizing FL in the mouse embryo.
Collapse
Affiliation(s)
- Daisuke Sugiyama
- Developmental Biology Laboratory, UPMC, CNRS UMR76229, 75252 Paris Cedex 05 France.
| | | | | |
Collapse
|
38
|
Cioffi DL, Wu S, Alexeyev M, Goodman SR, Zhu MX, Stevens T. Activation of the endothelial store-operated ISOC Ca2+ channel requires interaction of protein 4.1 with TRPC4. Circ Res 2005; 97:1164-72. [PMID: 16254212 DOI: 10.1161/01.res.0000193597.65217.00] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Store-operated calcium (SOC) entry represents the principal Ca2+ entry pathway into nonexcitable cells. Despite intensive investigation, mechanisms underlying activation of SOC entry have remained elusive. The endothelial ISOC channel is a Ca2+-selective SOC entry channel to which the transient receptor potential (TRP) proteins TRPC1 and TRPC4 contribute subunits. Activation of ISOC is specifically regulated by the spectrin-actin membrane skeleton; however, the nature of coupling between the ISOC channel and membrane skeleton is unknown. Here we demonstrate that protein 4.1 is an essential component of the ISOC channel gating mechanism. Protein 4.1 interacts with TRPC4 and the membrane skeleton. Deletion of the protein 4.1 binding domain on TRPC4 or peptide competition to the protein 4.1 binding domain prevents ISOC activation. These findings reveal that interaction of protein 4.1 with TRPC4 is required for activation of the endothelial ISOC channel.
Collapse
Affiliation(s)
- Donna L Cioffi
- Center for Lung Biology, Department of Pharmacology, University of South Alabama College of Medicine, Mobile, AL 36688, USA
| | | | | | | | | | | |
Collapse
|
39
|
Alvarez DF, King JA, Townsley MI. Resistance to store depletion-induced endothelial injury in rat lung after chronic heart failure. Am J Respir Crit Care Med 2005; 172:1153-60. [PMID: 16051904 PMCID: PMC2718399 DOI: 10.1164/rccm.200506-847oc] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
RATIONALE In chronic heart failure, the lung endothelial permeability response to angiotensin II or thapsigargin-induced store depletion is ablated, although the mechanisms are not understood. OBJECTIVES To determine whether the ablated permeability response to store depletion during heart failure was due to impaired expression of store operated Ca2+ channels in lung endothelium. METHODS Heart failure was induced by aortocaval fistula in rats. Permeability was measured in isolated lungs using the filtration coefficient and a low Ca2+/Ca2+ add-back strategy to identify the component of the permeability response dependent on Ca2+ entry. MAIN RESULTS In fistulas, right ventricular mass and left ventricular end diastolic pressure were increased and left ventricular shortening fraction decreased compared with shams. Thapsigargin-induced store depletion increased lung endothelial permeability in shams, but not in fistulas. Permeability increased in both groups after the Ca2+ ionophore A23187 or 14,15-epoxyeicosatrienoic acid, independent of store depletion. A diacylglycerol analog had no impact on permeability. Increased distance between the endoplasmic reticulum and the plasmalemmal membrane was ruled out as a mechanism for the loss of the permeability response to store depletion. Endothelial expression of the endoplasmic reticulum Ca2+ ATPase was not altered in fistulas compared with shams, whereas the store-operated canonical transient receptor potential channels 1, 3, and 4 were downregulated in extraalveolar vessel endothelium. CONCLUSIONS We conclude that the adaptive mechanism limiting store depletion-induced endothelial lung injury in the aortocaval model of heart failure involves downregulation of store-operated Ca2+ channels.
Collapse
Affiliation(s)
- Diego F Alvarez
- Department of Physiology, MSB 3074, University of South Alabama, Mobile, AL 36688, USA
| | | | | |
Collapse
|
40
|
Abstract
In electrically nonexcitable cells, Ca2+influx is essential for regulating a host of kinetically distinct processes involving exocytosis, enzyme control, gene regulation, cell growth and proliferation, and apoptosis. The major Ca2+entry pathway in these cells is the store-operated one, in which the emptying of intracellular Ca2+stores activates Ca2+influx (store-operated Ca2+entry, or capacitative Ca2+entry). Several biophysically distinct store-operated currents have been reported, but the best characterized is the Ca2+release-activated Ca2+current, ICRAC. Although it was initially considered to function only in nonexcitable cells, growing evidence now points towards a central role for ICRAC-like currents in excitable cells too. In spite of intense research, the signal that relays the store Ca2+content to CRAC channels in the plasma membrane, as well as the molecular identity of the Ca2+sensor within the stores, remains elusive. Resolution of these issues would be greatly helped by the identification of the CRAC channel gene. In some systems, evidence suggests that store-operated channels might be related to TRP homologs, although no consensus has yet been reached. Better understood are mechanisms that inactivate store-operated entry and hence control the overall duration of Ca2+entry. Recent work has revealed a central role for mitochondria in the regulation of ICRAC, and this is particularly prominent under physiological conditions. ICRACtherefore represents a dynamic interplay between endoplasmic reticulum, mitochondria, and plasma membrane. In this review, we describe the key electrophysiological features of ICRACand other store-operated Ca2+currents and how they are regulated, and we consider recent advances that have shed insight into the molecular mechanisms involved in this ubiquitous and vital Ca2+entry pathway.
Collapse
Affiliation(s)
- Anant B Parekh
- Department of Physiology, University of Oxford, United Kingdom.
| | | |
Collapse
|
41
|
Wu S, Cioffi EA, Alvarez D, Sayner SL, Chen H, Cioffi DL, King J, Creighton JR, Townsley M, Goodman SR, Stevens T. Essential role of a Ca2+-selective, store-operated current (ISOC) in endothelial cell permeability: determinants of the vascular leak site. Circ Res 2005; 96:856-63. [PMID: 15790951 DOI: 10.1161/01.res.0000163632.67282.1f] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Store-operated calcium (SOC) entry is sufficient to disrupt the extra-alveolar, but not the alveolar, endothelial cell barrier. Mechanism(s) underlying such insensitivity to transitions in cytosolic calcium ([Ca2+]i) in microvascular endothelial cells are unknown. Depletion of stored Ca2+ activates a larger SOC entry response in extra-alveolar (pulmonary artery; PAECs) than alveolar (pulmonary microvascular; PMVECs) endothelial cells. In vivo permeation studies revealed that Ca2+ store depletion activates similar nonselective cationic conductances in PAECs and PMVECs, while only PAECs possess the calcium-selective, store-operated Ca2+ entry current, I(SOC). Pretreatment with the type 4 phosphodiesterase inhibitor, rolipram, abolished thapsigargin-activated I(SOC) in PAECs, and revealed I(SOC) in PMVECs. Rolipram pretreatment shifted the thapsigargin-induced fluid leak site from extra-alveolar to alveolar vessels in the intact pulmonary circulation. Thus, our results indicate I(SOC) provides a [Ca2+]i source that is needed to disrupt the endothelial cell barrier, and demonstrate that intracellular events controlling I(SOC) activation coordinate the site-specific vascular response to inflammation.
Collapse
Affiliation(s)
- Songwei Wu
- Center for Lung Biology, The University of South Alabama College of Medicine, Mobile, Ala 36688, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
King J, Hamil T, Creighton J, Wu S, Bhat P, McDonald F, Stevens T. Structural and functional characteristics of lung macro- and microvascular endothelial cell phenotypes. Microvasc Res 2004; 67:139-51. [PMID: 15020205 DOI: 10.1016/j.mvr.2003.11.006] [Citation(s) in RCA: 175] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2003] [Indexed: 01/04/2023]
Abstract
Lung macro- and microvascular endothelial cells exhibit unique functional attributes, including signal transduction and barrier properties. We therefore sought to identify structural and functional features of endothelial cells that discriminate their phenotypes in the fully differentiated lung. Rat lung macro- (PAEC) and microvascular (PMVEC) endothelial cells each exhibited expression of typical markers. Screening for reactivity with nine different lectins revealed that Glycine max and Griffonia (Bandeiraea) simplicifolia preferentially bound microvascular endothelia whereas Helix pomatia preferentially bound macrovascular endothelia. Apposition between the apical plasmalemma and endoplasmic reticulum was closer in PAECs (8 nm) than in PMVECs (87 nm), implicating this coupling distance in the larger store operated calcium entry responses observed in macrovascular cells. PMVECs exhibited a faster growth rate than did PAECs and, once a growth program was initiated by serum, PMVECs sustained growth in the absence of serum. Thus, PAECs and PMVECs differ in their structure and function, even under similar environmental conditions.
Collapse
Affiliation(s)
- Judy King
- Department of Pathology, Center for Lung Biology, The University of South Alabama College of Medicine, Mobile, AL 36617, USA
| | | | | | | | | | | | | |
Collapse
|
43
|
Winter MC, Shasby SS, Ries DR, Shasby DM. Histamine selectively interrupts VE-cadherin adhesion independently of capacitive calcium entry. Am J Physiol Lung Cell Mol Physiol 2004; 287:L816-23. [PMID: 15220112 DOI: 10.1152/ajplung.00056.2004] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Histamine is an important agent of innate immunity, transiently increasing the flux of immune-competent molecules from the vascular space to the tissues and then allowing rapid restoration of the integrity of the endothelial barrier. In previous work we found that histamine alters the endothelial barrier by disrupting cell-cell adhesion and identified VE-cadherin as an essential participant in this process. The previous work did not determine whether histamine directly interrupted VE-cadherin adhesion, whether the effects of histamine were selective for cadherin adhesion, or whether capacitive calcium flux across the cell membrane was necessary for the effects of histamine on cell-cell adhesion. In the current work we found that histamine directly interrupts adhesion of L cells expressing the type 1 histamine (H1) receptor and VE-cadherin to a VE-cadherin-Fc fusion protein. In contrast, integrin-mediated adhesion to fibronectin of the same L cells expressing the H1 receptor was not affected by histamine, demonstrating that the effects of histamine are selective for cadherin adhesion. Some of the effects of many edemagenic agonists on endothelium are dependent on the capacitive flux of calcium across the endothelial cell membrane. Blocking capacitive calcium flux with LaCl3 did not prevent histamine from interrupting VE-cadherin adhesion of transfected L cells, nor did it prevent histamine from interrupting cell-cell adhesion of human umbilical vein endothelial cells. These data support the contentions that histamine directly and selectively interrupts cadherin adhesion and this effect on cadherin adhesion is independent of capacitive calcium flux.
Collapse
Affiliation(s)
- Michael C Winter
- Department of Internal Medicine, University of Iowa College of Medicine, Iowa City, Iowa 52242, USA
| | | | | | | |
Collapse
|
44
|
Alvarez DF, Gjerde EAB, Townsley MI. Role of EETs in regulation of endothelial permeability in rat lung. Am J Physiol Lung Cell Mol Physiol 2004; 286:L445-51. [PMID: 14578116 DOI: 10.1152/ajplung.00150.2003] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
This study tested the hypothesis that epoxyeicosatrienoic acids (EETs) derived from arachidonic acid via P-450 epoxygenases are soluble factors linking depletion of endoplasmic reticulum Ca2+ stores and store-dependent regulation of endothelial cell (EC) permeability in rat lung. EC permeability was measured via the capillary filtration coefficient ( Kf,c) in isolated, perfused rat lungs. 14,15-EET and 5,6-EET increased EC permeability, a response that was significantly different from that of 8,9-EET, 11,12-EET, and vehicle control. The permeability response to 14,15-EET was not significantly attenuated by the nonspecific Ca2+ channel blocker Gd3+ ( P = 0.068). In lungs perfused with low [Ca2+], 14,15-EET tended to increase EC permeability, although a significant increase in Kf,c was observed only following Ca2+ add-back. As positive control, we showed that the 3.7-fold increase in Kf,c evoked by thapsigargin (TG), a known activator of store depletion-induced Ca2+ entry, was blocked by both Gd3+ and low [Ca2+] buffer. Nonetheless, the permeability response to TG could not be blocked by the phospholipase A2 inhibitors mepacrine or methyl arachidonyl fluorophosphonate or the P-450 epoxygenase inhibitors 17-octadecynoic acid or propargyloxyphenyl hexanoic acid. Similarly, combined pretreatment with ibuprofen and dicyclohexylurea to block EET metabolism had no effect on the permeability response to TG. We conclude that EETs have a heterogeneous impact on EC permeability. Despite a requirement for Ca2+ entry with both TG and 14,15-EET, our data suggest that distinct signaling pathways or heterogeneity in EC responsiveness is responsible for the observed EC injury evoked by EETs and store depletion in the isolated rat lung.
Collapse
Affiliation(s)
- Diego F Alvarez
- Department of Physiology, University of Southern Alabama, Mobile, AL 36688, USA
| | | | | |
Collapse
|
45
|
Mehta D, Ahmmed GU, Paria BC, Holinstat M, Voyno-Yasenetskaya T, Tiruppathi C, Minshall RD, Malik AB. RhoA interaction with inositol 1,4,5-trisphosphate receptor and transient receptor potential channel-1 regulates Ca2+ entry. Role in signaling increased endothelial permeability. J Biol Chem 2003; 278:33492-500. [PMID: 12766172 DOI: 10.1074/jbc.m302401200] [Citation(s) in RCA: 171] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We tested the hypothesis that RhoA, a monomeric GTP-binding protein, induces association of inositol trisphosphate receptor (IP3R) with transient receptor potential channel (TRPC1), and thereby activates store depletion-induced Ca2+ entry in endothelial cells. We showed that RhoA upon activation with thrombin associated with both IP3R and TRPC1. Thrombin also induced translocation of a complex consisting of Rho, IP3R, and TRPC1 to the plasma membrane. IP3R and TRPC1 translocation and association required Rho activation because the response was not seen in C3 transferase (C3)-treated cells. Rho function inhibition using Rho dominant-negative mutant or C3 dampened Ca2+ entry regardless of whether Ca2+ stores were emptied by thrombin, thapsigargin, or inositol trisphosphate. Rho-induced association of IP3R with TRPC1 was dependent on actin filament polymerization because latrunculin (which inhibits actin polymerization) prevented both the association and Ca2+ entry. We also showed that thrombin produced a sustained Rho-dependent increase in cytosolic Ca2+ concentration [Ca2+]i in endothelial cells overexpressing TRPC1. We further showed that Rho-activated Ca2+ entry via TRPC1 is important in the mechanism of the thrombin-induced increase in endothelial permeability. In summary, Rho activation signals interaction of IP3R with TRPC1 at the plasma membrane of endothelial cells, and triggers Ca2+ entry following store depletion and the resultant increase in endothelial permeability.
Collapse
MESH Headings
- ADP Ribose Transferases/pharmacology
- Actins/chemistry
- Botulinum Toxins/pharmacology
- Calcium/metabolism
- Calcium Channels/chemistry
- Calcium Channels/metabolism
- Calcium Channels/physiology
- Cells, Cultured
- Electrophoresis, Polyacrylamide Gel
- Electrophysiology
- Endothelium, Vascular/cytology
- Endothelium, Vascular/metabolism
- Genes, Dominant
- Humans
- Inositol 1,4,5-Trisphosphate Receptors
- Microscopy, Confocal
- Models, Biological
- Patch-Clamp Techniques
- Precipitin Tests
- Protein Binding
- Protein Transport
- Receptors, Cytoplasmic and Nuclear/chemistry
- Receptors, Cytoplasmic and Nuclear/metabolism
- Signal Transduction
- TRPC Cation Channels
- Thapsigargin/chemistry
- Thapsigargin/pharmacology
- Thrombin/chemistry
- Time Factors
- Transfection
- rho GTP-Binding Proteins/metabolism
- rhoA GTP-Binding Protein/chemistry
- rhoA GTP-Binding Protein/metabolism
Collapse
Affiliation(s)
- Dolly Mehta
- Department of Pharmacology, College of Medicine, The University of Illinois, Chicago, Illinois 60612, USA.
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Sugiyama D, Ogawa M, Hirose I, Jaffredo T, Arai KI, Tsuji K. Erythropoiesis from acetyl LDL incorporating endothelial cells at the preliver stage. Blood 2003; 101:4733-8. [PMID: 12595314 DOI: 10.1182/blood-2002-09-2799] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Erythropoiesis is characterized by 2 waves of production during mouse embryogenesis: a primitive one originating from the yolk sac (YS) and a definitive one produced from both the YS and the embryo proper. How the latter wave is generated remains unclear. To investigate our hypothesis that endothelial cells (ECs) could generate erythroid cells, we designed a method to label ECs at 10 days after coitus. This labeling method associates 2 techniques: an intracardiac inoculation that allows molecules to be delivered into the bloodstream followed by a whole-embryo culture period. DiI-conjugated acetylated low-density lipoproteins (Ac-LDL-DiI) were used to specifically tag ECs from the inside. One hour after inoculation, DiI staining was found along the entire endothelial tree. Fluorescence-activated cell sorter (FACS) analysis revealed that DiI+ cells were CD31+, CD34+, and CD45-, an antigen makeup characteristic of the endothelial lineage. Twelve hours after inoculation, 43% of DiI+ circulating cells belonged to the erythroid lineage. These cells expressed Ter119 and displayed an adult globin chain arrangement; thus they belonged to the definitive lineage as confirmed in erythroid colony formation. The remaining cells likely represent committed white blood cells or multipotent progenitors, as revealed by a mixed-colony formation. Beyond the 29-somite stage, the proportion of DiI+ erythroid cells gradually decreased. These results demonstrate the generation of hematopoietic cells from an endothelial intermediate, using in vivo tracing. We provide evidence for a release of these cells into the circulation and hypothesize that these cells are able to colonize the fetal liver and generate definitive erythrocytes in vivo.
Collapse
Affiliation(s)
- Daisuke Sugiyama
- Division of Cellular Therapy, Advanced Clinical Research Center, Institute of Medical Science, University of Tokyo, Japan.
| | | | | | | | | | | |
Collapse
|
47
|
Abstract
Ca2+ store depletion activates both Ca2+ selective and non-selective currents in endothelial cells. Recently, considerable progress has been made in understanding the molecular make-up and regulation of an endothelial cell thapsigargin-activated Ca2+ selective current, I(SOC). Indeed, I(SOC) is a relatively small inward Ca2+ current that exhibits an approximate +40mV reversal potential and is strongly inwardly rectifying. This current is sensitive to organization of the actin-based cytoskeleton. Transient receptor potential (TRP) proteins 1 and 4 (TRPC1 and TRPC4, respectively) each contribute to the molecular basis of I(SOC), although it is TRPC4 that appears to be tethered to the cytoskeleton through a dynamic interaction with protein 4.1. Activation of I(SOC) requires association between protein 4.1 and the actin-based cytoskeleton (mediated through spectrin), suggesting protein 4.1 mediates the physical communication between Ca2+ store depletion and channel activation. Thus, at present findings indicate a TRPC4-protein 4.1 physical linkage regulates I(SOC) activation following Ca2+ store depletion.
Collapse
Affiliation(s)
- Donna L Cioffi
- Department of Pharmacology, Center for Lung Biology, University of South Alabama College of Medicine, Mobile, AL 36688, USA
| | | | | |
Collapse
|
48
|
Ramez M, Blot-Chabaud M, Cluzeaud F, Chanan S, Patterson M, Walensky LD, Marfatia S, Baines AJ, Chasis JA, Conboy JG, Mohandas N, Gascard P. Distinct distribution of specific members of protein 4.1 gene family in the mouse nephron. Kidney Int 2003; 63:1321-37. [PMID: 12631349 DOI: 10.1046/j.1523-1755.2003.00870.x] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
BACKGROUND Protein 4.1 is an adapter protein that links the actin cytoskeleton to various transmembrane proteins. These 4.1 proteins are encoded by four homologous genes, 4.1R, 4.1G, 4.1N, and 4.1B, which undergo complex alternative splicing. Here we performed a detailed characterization of the expression of specific 4.1 proteins in the mouse nephron. METHODS Distribution of renal 4.1 proteins was investigated by staining of paraformaldehyde-fixed mouse kidney sections with antibodies highly specific for each 4.1 protein. Major 4.1 splice forms, amplified from mouse kidney marathon cDNA, were expressed in transfected COS-7 cells in order to assign species of known exon composition to proteins detected in kidney. RESULTS A 105 kD 4.1R splice form, initiating at ATG-2 translation initiation site and lacking exon 16, but including exon 17B, was restricted to thick ascending limb of Henle's loop. A 95 kD 4.1N splice form, lacking exons 15 and 17D, was expressed in either descending or ascending thin limb of Henle's loop, distal convoluted tubule, and all regions of the collecting duct system. A major 108 kD 4.1B splice form, initiating at a newly characterized ATG translation initiation site, and lacking exons 15, 17B, and 21, was present only in Bowman's capsule and proximal convoluted tubule (PCT). There was no expression of 4.1G in kidney. CONCLUSION Distinct distribution of 4.1 proteins along the nephron suggests their involvement in targeting of selected transmembrane proteins in kidney epithelium and, therefore, in regulation of specific kidney functions.
Collapse
Affiliation(s)
- Mohamed Ramez
- Department of Subcellular Structure, Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Zhang S, Mizutani A, Hisatsune C, Higo T, Bannai H, Nakayama T, Hattori M, Mikoshiba K. Protein 4.1N is required for translocation of inositol 1,4,5-trisphosphate receptor type 1 to the basolateral membrane domain in polarized Madin-Darby canine kidney cells. J Biol Chem 2003; 278:4048-56. [PMID: 12444087 DOI: 10.1074/jbc.m209960200] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Protein 4.1N was identified as a binding molecule for the C-terminal cytoplasmic tail of inositol 1,4,5-trisphosphate receptor type 1 (IP(3)R1) using a yeast two-hybrid system. 4.1N and IP(3)R1 associate in both subconfluent and confluent Madin-Darby canine kidney (MDCK) cells, a well studied tight polarized epithelial cell line. In subconfluent MDCK cells, 4.1N is distributed in the cytoplasm and the nucleus; IP(3)R1 is localized in the cytoplasm. In confluent MDCK cells, both 4.1N and IP(3)R1 are predominantly translocated to the basolateral membrane domain, whereas 4.1R, the prototypical homologue of 4.1N, is localized at the tight junctions (Mattagajasingh, S. N., Huang, S. C., Hartenstein, J. S., and Benz, E. J., Jr. (2000) J. Biol. Chem. 275, 30573-30585), and other endoplasmic reticulum marker proteins are still present in the cytoplasm. Moreover, the 4.1N-binding region of IP(3)R1 is necessary and sufficient for the localization of IP(3)R1 at the basolateral membrane domain. A fragment of the IP(3)R1-binding region of 4.1N blocks the localization of co-expressed IP(3)R1 at the basolateral membrane domain. These data indicate that 4.1N is required for IP(3)R1 translocation to the basolateral membrane domain in polarized MDCK cells.
Collapse
Affiliation(s)
- Songbai Zhang
- Division of Molecular Neurobiology, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Venkatachalam K, van Rossum DB, Patterson RL, Ma HT, Gill DL. The cellular and molecular basis of store-operated calcium entry. Nat Cell Biol 2002; 4:E263-72. [PMID: 12415286 DOI: 10.1038/ncb1102-e263] [Citation(s) in RCA: 294] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The impact of calcium signalling on so many areas of cell biology reflects the crucial role of calcium signals in the control of diverse cellular functions. Despite the precision with which spatial and temporal details of calcium signals have been resolved, a fundamental aspect of the generation of calcium signals -- the activation of 'store-operated channels' (SOCs) -- remains a molecular and mechanistic mystery. Here we review new insights into the exchange of signals between the endoplasmic reticulum (ER) and plasma membrane that result in activation of calcium entry channels mediating crucial long-term calcium signals.
Collapse
Affiliation(s)
- Kartik Venkatachalam
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | | | | | | | | |
Collapse
|