12
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: not found
      • Article: not found

      Reduced membrane cholesterol limits pulmonary endothelial Ca2+ entry after chronic hypoxia

      Read this article at

      ScienceOpenPublisherPMC
      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          <p class="first" id="d9374621e169">This research is the first to examine the direct role of membrane cholesterol in regulating pulmonary endothelial agonist-induced Ca <sup>2+</sup> entry and its components. The results provide a potential mechanism by which chronic hypoxia impairs pulmonary endothelial Ca <sup>2+</sup> influx, which may contribute to pulmonary hypertension. </p><p class="first" id="d9374621e178">Chronic hypoxia (CH)-induced pulmonary hypertension is associated with diminished production of endothelium-derived Ca <sup>2+</sup>-dependent vasodilators such as nitric oxide. Interestingly, ATP-induced endothelial Ca <sup>2+</sup> entry as well as membrane cholesterol (Chol) are decreased in pulmonary arteries from CH rats (4 wk, barometric pressure = 380 Torr) compared with normoxic controls. Store-operated Ca <sup>2+</sup> entry (SOCE) and depolarization-induced Ca <sup>2+</sup> entry are major components of the response to ATP and are similarly decreased after CH. We hypothesized that membrane Chol facilitates both SOCE and depolarization-induced pulmonary endothelial Ca <sup>2+</sup> entry and that CH attenuates these responses by decreasing membrane Chol. To test these hypotheses, we administered Chol or epicholesterol (Epichol) to acutely isolated pulmonary arterial endothelial cells (PAECs) from control and CH rats to either supplement or replace native Chol, respectively. The efficacy of membrane Chol manipulation was confirmed by filipin staining. Epichol greatly reduced ATP-induced Ca <sup>2+</sup> influx in PAECs from control rats. Whereas Epichol similarly blunted endothelial SOCE in PAECs from both groups, Chol supplementation restored diminished SOCE in PAECs from CH rats while having no effect in controls. Similar effects of Chol manipulation on PAEC Ca <sup>2+</sup> influx were observed in response to a depolarizing stimulus of KCl. Furthermore, KCl-induced Ca <sup>2+</sup> entry was inhibited by the T-type Ca <sup>2+</sup> channel antagonist mibefradil but not the L-type Ca <sup>2+</sup> channel inhibitor diltiazem. We conclude that PAEC membrane Chol is required for ATP-induced Ca <sup>2+</sup> entry and its two components, SOCE and depolarization-induced Ca <sup>2+</sup> entry, and that reduced Ca <sup>2+</sup> entry after CH may be due to loss of this key regulator. </p><p id="d9374621e221"> <b>NEW &amp; NOTEWORTHY</b> This research is the first to examine the direct role of membrane cholesterol in regulating pulmonary endothelial agonist-induced Ca <sup>2+</sup> entry and its components. The results provide a potential mechanism by which chronic hypoxia impairs pulmonary endothelial Ca <sup>2+</sup> influx, which may contribute to pulmonary hypertension. </p>

          Related collections

          Most cited references53

          • Record: found
          • Abstract: found
          • Article: not found

          Use of cyclodextrins to manipulate plasma membrane cholesterol content: evidence, misconceptions and control strategies.

          The physiological importance of cholesterol in the cell plasma membrane has attracted increased attention in recent years. Consequently, the use of methods of controlled manipulation of membrane cholesterol content has also increased sharply, especially as a method of studying putative cholesterol-enriched cell membrane domains (rafts). The most common means of modifying the cholesterol content of cell membranes is the incubation of cells or model membranes with cyclodextrins, a family of compounds, which, due to the presence of relatively hydrophobic cavity, can be used to extract cholesterol from cell membranes. However, the mechanism of this activity of cyclodextrins is not completely established. Moreover, under conditions commonly used for cholesterol extraction, cyclodextrins may remove cholesterol from both raft and non-raft domains of the membrane as well as alter the distribution of cholesterol between plasma and intracellular membranes. In addition, other hydrophobic molecules such as phospholipids may also be extracted from the membranes by cyclodextrins. We review the evidence for the specific and non-specific effects of cyclodextrins and what is known about the mechanisms for cyclodextrin-induced cholesterol and phospholipid extraction. Finally, we discuss useful control strategies that may help to verify that the observed effects are due specifically to cyclodextrin-induced changes in cellular cholesterol.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Expression of angiogenesis-related molecules in plexiform lesions in severe pulmonary hypertension: evidence for a process of disordered angiogenesis.

            Pulmonary arteries of patients with severe pulmonary hypertension (SPH) presenting in an idiopathic form (primary PH-PPH) or associated with congenital heart malformations or collagen vascular diseases show plexiform lesions. It is postulated that in lungs with SPH, endothelial cells in plexiform lesions express genes encoding for proteins involved in angiogenesis, in particular, vascular endothelial growth factor (VEGF) and those involved in VEGF receptor-2 (VEGFR-2) signalling. On immunohistochemistry and in situ hybridization, endothelial cells in the plexiform lesions expressed VEGF mRNA and protein and overexpressed the mRNA and protein of VEGFR-2, and the transcription factor subunits HIF-1alpha and HIF-1beta of hypoxia inducible factor, which are responsible for the hypoxia-dependent induction of VEGF. When compared with normal lungs, SPH lungs showed decreased expression of the kinases PI3 kinase and src, which, together with Akt, relay the signal transduction downstream of VEGFR-2. Because markers of angiogenesis are expressed in plexiform lesions in SPH, it is proposed that these lesions may form by a process of disordered angiogenesis. Copyright 2001 John Wiley & Sons, Ltd.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Stim1 and Orai1 mediate CRAC currents and store-operated calcium entry important for endothelial cell proliferation.

              Recent breakthroughs in the store-operated calcium (Ca(2+)) entry (SOCE) pathway have identified Stim1 as the endoplasmic reticulum Ca(2+) sensor and Orai1 as the pore forming subunit of the highly Ca(2+)-selective CRAC channel expressed in hematopoietic cells. Previous studies, however, have suggested that endothelial cell (EC) SOCE is mediated by the nonselective canonical transient receptor potential channel (TRPC) family, TRPC1 or TRPC4. Here, we show that passive store depletion by thapsigargin or receptor activation by either thrombin or the vascular endothelial growth factor activates the same pathway in primary ECs with classical SOCE pharmacological features. ECs possess the archetypical Ca(2+) release-activated Ca(2+) current (I(CRAC)), albeit of a very small amplitude. Using a maneuver that amplifies currents in divalent-free bath solutions, we show that EC CRAC has similar characteristics to that recorded from rat basophilic leukemia cells, namely a similar time course of activation, sensitivity to 2-aminoethoxydiphenyl borate, and low concentrations of lanthanides, and large Na(+) currents displaying the typical depotentiation. RNA silencing of either Stim1 or Orai1 essentially abolished SOCE and I(CRAC) in ECs, which were rescued by ectopic expression of either Stim1 or Orai1, respectively. Surprisingly, knockdown of either TRPC1 or TRPC4 proteins had no effect on SOCE and I(CRAC). Ectopic expression of Stim1 in ECs increased their I(CRAC) to a size comparable to that in rat basophilic leukemia cells. Knockdown of Stim1, Stim2, or Orai1 inhibited EC proliferation and caused cell cycle arrest at S and G2/M phase, although Orai1 knockdown was more efficient than that of Stim proteins. These results are first to our knowledge to establish the requirement of Stim1/Orai1 in the endothelial SOCE pathway.
                Bookmark

                Author and article information

                Journal
                American Journal of Physiology-Heart and Circulatory Physiology
                American Journal of Physiology-Heart and Circulatory Physiology
                American Physiological Society
                0363-6135
                1522-1539
                June 2017
                June 2017
                : 312
                : 6
                : H1176-H1184
                Article
                10.1152/ajpheart.00097.2017
                5495930
                28364016
                458e8183-4c8f-4bf2-a61f-b2aa7fcc0711
                © 2017
                History

                Comments

                Comment on this article