Skip to main content

OPINION article

Front. Med., 04 September 2023
Sec. Translational Medicine
This article is part of the Research Topic Molecular Mechanisms of Lung Endothelial Permeability, Vol II View all 4 articles

Heat shock protein 90 inhibition in the endothelium

  • School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, LA, United States

Introduction

Heat shock protein 90 (Hsp90) is a molecular chaperone assisting in the folding and maturation of a plethora of intracellular proteins, which participate in crucial functions and responses, including inflammation (1). Hsp90 inhibitors were developed—and are tested in clinical trials—to oppose cancers; and have been associated with anti-inflammatory activities (2). Those effects are not limited to malignant tissues, but are also applied in endothelial cells (35).

Endothelial inflammation promotes barrier dysfunction, tissue leak, lung edema, which are considered the hallmarks of acute respiratory distress syndrome (6). This is a respiratory disorder, associated with high mortality rates in the intensive care units, in septic patients. The COVID-19—related ARDS has caused more than 1,100,000 deaths in the Unites States (7), and efficient medicine to counteract it does not exist, so far.

Blocking the COVID-19—related cytokine storm it is a promising therapeutic strategy, and anti-inflammatory agents appear to improve patient survival. However, glucocorticoids are not efficient in cases of substantial inflammation, and monoclonal antibodies were developed to suppress the cytokine storm (8). IL-1 blockade delivered promising results (9). Indeed, there is an urgent need to develop new therapeutics against ARDS, utilizing robust anti-inflammatory agents.

Hsp90 inhibitors represent a promising therapeutic approach to oppose lung inflammatory disease, so to reinstate normal endothelial barrier function (10). In addition to their ability to block transcriptions factors which propel inflammatory responses (e.g., NFκB) (11), they can induce survival elements in charge of cell homeostasis, to ameliorate injury. P53 participates in those events.

P53 is a transcription factor which opposes the activities of NFkB in human tissues (12), and P53 deletion worsens LPS-induced injury in mice (13). P53 inhibition using pifithrin or small interfering RNA potentiated endothelial inflammation, while P53 augmentation exerted protective effects (14). The guardian of the genome mediates—at least in part—the effects of Hsp90 inhibition in the lungs, and mice overexpressing P53 were protected against inflammatory lung injury (15). Moreover, Hsp90 inhibition suppresses P53 phosphorylation, preventing P53 degradation (16, 17). The actin cytoskeleton is affected by P53, since this transcription factor can induce cortical actin, and suppress filamentous actin formation (18). The unfolded protein response (UPR) can also participate in the Hsp90 inhibitors—related effects in the endothelium.

UPR is a mechanism involved in cell fate, and can initiate repairing processes or induce cell death (19, 20). It is involved in endothelial barrier function. Recent studies suggest that UPR activation increases barrier integrity and reduces endothelial permeability, whereas its suppression is associated to impaired barrier function (2125). Hsp90 inhibitors were shown to activate UPR sensors, as well as their downstream targets, in endothelial cells and mouse lungs (26, 27). The effects of Hsp90 inhibitors are also applied to brain microvascular cells, a component of the blood brain barrier. Specifically, those compounds protect brain cells against LPS (28) and oxidative stress (28, 29); in line with similar P53—mediated effects, in vitro (30, 31).

Hsp90 inhibitors may represent an exciting new possibility to counteract COVID-19. SARS-CoV-2 spike triggers barrier dysfunction and vascular leak via integrins and TGF-β signaling (32). The aforementioned compounds modulate SARS-CoV-2 spike protein subunit 1-induced human pulmonary microvascular endothelial activation and barrier dysfunction (33). They can also suppress SARS-CoV-2 assembly partially through induced M or N degradation (34). Interestingly, the oral Hsp90 inhibitor SNX-5422 attenuates SARS-CoV-2 replication and suppresses inflammation in airway cells (35).

Discussion

Many questions are to be addressed about the specific mechanisms by which Hsp90 inhibition assists impaired/inflamed endothelial cells to survive, and affected tissues to recover. Which are the exact kinases mediating the effects of Hsp90 inhibitors toward P53 modulation, and how this molecular chaperone modulates UPR in endothelial cells? It was previous reported that IRE1α is involved in those phenomena, in cancers (36). Studies in genetically modified mice which do not express P53 and UPR sensors in their lung endothelium will most probably address those questions; to enrich our knowledge on the expanding Hsp90 universe.

Author contributions

NB: Writing—original draft, Writing—review and editing.

Funding

The author declares financial support was received for the research, authorship, and/or publication of this article. NB's research is supported by the i) R&D, Research Competitiveness Subprogram of the Louisiana Board of Regents through the Board of Regents Support Fund [LEQSF (2019–2022)-RD-A-26] and ii) Institutional Development Award (IDeA) from the National Institute of General Medical Sciences of the National Institutes of Health under grant No. P2O GM103424-21.

Conflict of interest

The author declares that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

The author declared that they were an editorial board member of Frontiers, at the time of submission. This had no impact on the peer review process and the final decision.

Publisher's note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

1. Barabutis N, Verin A, Catravas JD. Regulation of pulmonary endothelial barrier function by kinases. Am J Physiol Lung Cell Mol Physiol. (2016) 311:L832–45. doi: 10.1152/ajplung.00233.2016

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Dong B, Jaeger AM, Thiele DJ. Inhibiting heat shock factor 1 in cancer: a unique therapeutic opportunity. Trends Pharmacol Sci. (2019) 40:986–1005. doi: 10.1016/j.tips.2019.10.008

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Chatterjee A, Dimitropoulou C, Drakopanayiotakis F, Antonova G, Snead C, Cannon J, et al. Heat shock protein 90 inhibitors prolong survival, attenuate inflammation, and reduce lung injury in murine sepsis. Am J Respir Crit Care Med. (2007) 176:667–75. doi: 10.1164/rccm.200702-291OC

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Antonov A, Snead C, Gorshkov B, Antonova GN, Verin AD, Catravas JD. Heat shock protein 90 inhibitors protect and restore pulmonary endothelial barrier function. Am J Respir Cell Mol Biol. (2008) 39:551–9. doi: 10.1165/rcmb.2007-0324OC

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Chatterjee A, Snead C, Yetik-Anacak G, Antonova G, Zeng J, Catravas JD. Heat shock protein 90 inhibitors attenuate LPS-induced endothelial hyperpermeability. Am J Physiol Lung Cell Mol Physiol. (2008) 294:L755–63. doi: 10.1152/ajplung.00350.2007

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Siejka A, Barabutis N. Adrenal insufficiency in the COVID-19 era. Am J Physiol Endocrinol Metab. (2021) 320:E784–5. doi: 10.1152/ajpendo.00061.2021

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Pozzi T, Collino F, Brusatori S, Romitti F, Busana M, Moerer O, et al. Specific respiratory system compliance in COVID-19 and Non-COVID-19 acute respiratory distress syndrome. Am J Respir Crit Care Med. (2023) 208:328–30. doi: 10.1164/rccm.202302-0223LE

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Cron RQ, Caricchio R, Chatham WW. Calming the cytokine storm in COVID-19. Nat Med. (2021) 27:1674–5. doi: 10.1038/s41591-021-01500-9

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Kyriazopoulou E, Poulakou G, Milionis H, Metallidis S, Adamis G, Tsiakos K, et al. Early treatment of COVID-19 with anakinra guided by soluble urokinase plasminogen receptor plasma levels: a double-blind, randomized controlled phase 3 trial. Nat Med. (2021) 27:1752–60. doi: 10.1038/s41591-021-01499-z

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Barabutis N. Heat shock protein 90 inhibition in the inflamed lungs. Cell Stress Chaperones. (2020) 25:195–7. doi: 10.1007/s12192-020-01069-1

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Thangjam GS, Birmpas C, Barabutis N, Gregory BW, Clemens MA, Newton JR, et al. Hsp90 inhibition suppresses NF-kappaB transcriptional activation via Sirt-2 in human lung microvascular endothelial cells. Am J Physiol Lung Cell Mol Physiol. (2016) 310:L964–74. doi: 10.1152/ajplung.00054.2016

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Uddin MA, Barabutis N. P53 in the impaired lungs. DNA Repair (Amst). (2020) 95:102952. doi: 10.1016/j.dnarep.2020.102952

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Uddin MA, Akhter MS, Kubra K-T, Barabutis N. P53 deficiency potentiates LPS-Induced acute lung injury in vivo. Curr Res Physiol. (2020) 3:30–3. doi: 10.1016/j.crphys.2020.07.001

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Barabutis N, Dimitropoulou C, Birmpas C, Joshi A, Thangjam G, Catravas JD. p53 protects against LPS-induced lung endothelial barrier dysfunction. Am J Physiol Lung Cell Mol Physiol. (2015) 308:L776–87. doi: 10.1152/ajplung.00334.2014

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Barabutis N, Dimitropoulou C, Gregory B, Catravas JD. Wild-type p53 enhances endothelial barrier function by mediating RAC1 signalling and RhoA inhibition. J Cell Mol Med. (2018) 22:1792–804. doi: 10.1111/jcmm.13460

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Kubra KT, Barabutis N. P53 in endothelial function and unfolded protein response regulation. Cell Biol Int. (2022) 46:2257–61. doi: 10.1002/cbin.11891

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Barabutis N, Uddin MA, Catravas JD. Hsp90 inhibitors suppress P53 phosphorylation in LPS - induced endothelial inflammation. Cytokine. (2019) 113:427–32. doi: 10.1016/j.cyto.2018.10.020

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Barabutis N. P53 in RhoA regulation. Cytoskeleton (Hoboken). (2020) 77:197–201. doi: 10.1002/cm.21604

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Kubra K-T, Akhter MS, Uddin MA, Barabutis N. Unfolded protein response in cardiovascular disease. Cell Signal. (2020) 73:109699. doi: 10.1016/j.cellsig.2020.109699

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Akhter MS, Uddin MA, Kubra KT, Barabutis N. Autophagy, unfolded protein response and lung disease. Curr Res Cell Biol. (2020) 1:100003. doi: 10.1016/j.crcbio.2020.100003

PubMed Abstract | CrossRef Full Text | Google Scholar

21. Kubra KT, Barabutis N. Brefeldin A and kifunensine modulate LPS-induced lung endothelial hyperpermeability in human and bovine cells. Am J Physiol Cell Physiol. (2021) 321:C214–20. doi: 10.1152/ajpcell.00142.2021

PubMed Abstract | CrossRef Full Text | Google Scholar

22. Akhter MS, Kubra K-T, Uddin MA, Barabutis N. Kifunensine compromises lung endothelial barrier function. Microvasc Res. (2020) 132:104051. doi: 10.1016/j.mvr.2020.104051

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Kubra K-T, Uddin MA, Akhter MS, Barabutis N. Luminespib counteracts the Kifunensine-induced lung endothelial barrier dysfunction. Curr Res Toxicol. (2020) 1:111–5. doi: 10.1016/j.crtox.2020.09.003

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Barabutis N, Akhter MS. Unfolded protein response suppression potentiates LPS-induced barrier dysfunction and inflammation in bovine pulmonary artery endothelial cells. Tissue Barriers. (2023) 12:2232245. doi: 10.1080/21688370.2023.2232245

PubMed Abstract | CrossRef Full Text | Google Scholar

25. Kubra KT, Barabutis N. Ceapin-A7 potentiates lipopolysaccharide-induced endothelial injury. J Biochem. Mol Toxicol. (2023) 11:e23460. doi: 10.1002/jbt.23460

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Uddin MA, Kubra K-T, Sonju JJ, Akhter MS, Seetharama J, Barabutis N. Effects of heat shock protein 90 inhibition in the lungs. Med Drug Discov. (2020) 6:100046. doi: 10.1016/j.medidd.2020.100046

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Kubra K-T, Uddin MA, Akhter MS, Barabutis N. Hsp90 inhibitors induce the unfolded protein response in bovine and mice lung cells. Cell Signal. (2020) 67:109500. doi: 10.1016/j.cellsig.2019.109500

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Uddin MA, Akhter MS, Kubra K-T, Barabutis N. Hsp90 inhibition protects brain endothelial cells against LPS-induced injury. Biofactors. (2022) 48:926–33. doi: 10.1002/biof.1833

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Uddin MA, Akhter MS, Kubra K-T, Whitaker KE, Shipley SL, Smith LM, et al. Hsp90 inhibition protects the brain microvascular endothelium against oxidative stress. Brain Disord. (2021) 1:100001. doi: 10.1016/j.dscb.2020.100001

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Akhter MS, Uddin MA, Kubra KT, Barabutis N. P53-induced reduction of lipid peroxidation supports brain microvascular endothelium integrity. J Pharmacol Sci. (2019) 141:83–5. doi: 10.1016/j.jphs.2019.09.008

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Barabutis N. Insights on supporting the aging brain microvascular endothelium. Aging Brain. (2021) 1:100009. doi: 10.1016/j.nbas.2021.100009

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Biering SB, Gomes de Sousa FT, Tjang LV, Pahmeier F, Zhu C, Ruan R, et al. SARS-CoV-2 Spike triggers barrier dysfunction and vascular leak via integrins and TGF-beta signaling. Nat Commun. (2022) 13:7630. doi: 10.1038/s41467-022-34910-5

PubMed Abstract | CrossRef Full Text | Google Scholar

33. Biancatelli RMLC, Solopov PA, Gregory B, Khodour Y, Catravas JD. HSP90 inhibitors modulate SARS-CoV-2 spike protein subunit 1-induced human pulmonary microvascular endothelial activation and barrier dysfunction. Front Physiol. (2022) 13:812199. doi: 10.3389/fphys.2022.812199

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Zhao Z, Xu L-D, Zhang F, Liang Q-Z, Jiao Y, Shi F-S, et al. Heat shock protein 90 facilitates SARS-CoV-2 structural protein-mediated virion assembly and promotes virus-induced pyroptosis. J Biol Chem. (2023) 299:104668. doi: 10.1016/j.jbc.2023.104668

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Goswami R, Russell VS, Tu JJ, Thomas C, Hughes P, Kelly F, et al. Oral Hsp90 inhibitor SNX-5422 attenuates SARS-CoV-2 replication and dampens inflammation in airway cells. iScience. (2021) 24:103412. doi: 10.1016/j.isci.2021.103412

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Marcu MG, Doyle M, Bertolotti A, Ron D, Hendershot L, Neckers L. Heat shock protein 90 modulates the unfolded protein response by stabilizing IRE1alpha. Mol Cell Biol. (2002) 22:8506–13. doi: 10.1128/MCB.22.24.8506-8513.2002

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: inflammation, lung injury, unfolded protein response, acute respiratory distress syndrome, COVID-19

Citation: Barabutis N (2023) Heat shock protein 90 inhibition in the endothelium. Front. Med. 10:1255488. doi: 10.3389/fmed.2023.1255488

Received: 08 July 2023; Accepted: 23 August 2023;
Published: 04 September 2023.

Edited by:

Alexander Verin, Augusta University, United States

Reviewed by:

Pavel Solopov, Old Dominion University, United States

Copyright © 2023 Barabutis. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Nektarios Barabutis, barabutis@ulm.edu

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.