167
Views
3
CrossRef citations to date
0
Altmetric
Brief Report

Unfolded protein response suppression potentiates LPS-induced barrier dysfunction and inflammation in bovine pulmonary artery endothelial cells

&
Article: 2232245 | Received 22 Apr 2023, Accepted 27 Jun 2023, Published online: 12 Jul 2023

ABSTRACT

The development of novel strategies to counteract diseases related to barrier dysfunction is a priority, since sepsis and acute respiratory distress syndrome are still associated with high mortality rates. In the present study, we focus on the effects of the unfolded protein response suppressor (UPR) 4-Phenylbutyrate (4-PBA) in Lipopolysaccharides (LPS)-induced endothelial injury, to investigate the effects of that compound in the corresponding damage. 4-PBA suppressed binding immunoglobulin protein (BiP) – a UPR activation marker – and potentiated LPS – induced signal transducer and activator of transcription 3 (STAT3) and extracellular signal‑regulated protein kinase (ERK) 1/2 activation. In addition to those effects, 4-PBA enhanced paracellular hyperpermeability in inflamed bovine pulmonary endothelial cells, and did not affect cell viability in moderate concentrations. Our observations suggest that UPR suppression due to 4-PBA augments LPS-induced endothelial injury, as well as the corresponding barrier disruption.

Introduction

Organ function relies on blood vessel permeability, which adapts to physiological needs.Citation1 The vascular bed of pulmonary, coronary, and skeletal muscle is composed of endothelial cells, forming a restrictive barrier, the vascular endothelium. This is a semi-permeable lining of blood vessels, formed to control the extravasation of nutrients, proteins, and electrolytes to the underlying tissues. Its integrity is important for crucial physiological conditions, including tissue-fluid homeostasis, vascular tone, host defense, and angiogenesis.Citation2 Endothelial leakage can impair gas exchange; leading to hypoxia, hypercapnia, acute lung injury (ALI), or acute respiratory distress syndrome ARDS.Citation3 Targeted medicine for ARDS does not exist, hence the mortality rates of the affected individuals are unacceptably high; as evident in the recent pandemic.Citation4

Endoplasmic reticulum (ER) accommodates a wide range of chaperones and enzymes for protein maturation. Hypoxia, redox changes, increased secretory load, and nutrient deprivation disrupt ER homeostasis, leading to deposition of misfolded or unfolded proteins in the ER lumen.Citation5 Those events activate one or more unfolded protein response (UPR) sensors; namely the protein kinase RNA like ER kinase (PERK), activating transcription factor 6 (ATF6), and inositol-requiring enzyme 1α (IRE1α). In unstressed conditions, those protein-sensors are attached to the binding immunoglobulin protein (BiP). It has been suggested that UPR is involved in endothelial repairing processes,Citation6 and may represent a therapeutic target for ARDS.Citation7 UPR modulation appears to hold the capacity to affect barrier function,Citation8,Citation9 but more information on that topic is needed.

The effects of UPR suppressor 4-phenylbutyric acid (4-PBA) in LPS-induced signal transducer and activator of transcription 3 (STAT3), extracellular signal-regulated kinase 2 (ERK2) activation; as well as in the paracellular permeability of lung endothelial cells have not been investigated yet. Lipopolysaccharides activates Toll-like receptor 4 (TLR-4 receptor), to induce inflammation, and has been extensively used to inflict lung injury in vitro, as well as in vivo.Citation10,Citation11 4-PBA is a chemical chaperone which prevents the aggregation of misfolded proteins and reduces endoplasmic reticulum stress.Citation12

Herein, we report for the first time that 4-PBA potentiates LPS-induced endothelial inflammation in bovine pulmonary artery endothelial cells (BPAEC); as well as the corresponding paracellular hyperpermeability. Moderate doses (0.1–20 mM, 24 hours) of that compound did not affect cell viability. Our study contributes in our understanding of the UPR-related actions in the endothelial context, and suggest that targeting UPR might be a promising pharmacological approach to strengthen barrier function. Future in vivo studies will further explore the potential of the aforementioned possibility.

Materials and methods

Reagents

RIPA buffer (AAJ63306-AP), anti-mouse IgG HRP-linked antibody (95017–554), anti-rabbit IgG HRP-linked antibody (95017–556), nitrocellulose membranes (10063–173), and 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) (BT142015-5 G) were purchased from VWR (Radnor, PA, USA). The BiP (3183S), phospho-STAT3 (9145S), STAT3 (4904S), phospho-p44/42 MAPK (ERK1/2) (9101S), and p44/42 MAPK (ERK1/2) (9102S) antibodies were obtained from Cell Signaling Technology (Danvers, MA, USA). The β-actin antibody (A5441), lipopolysaccharides (LPS) (L4130), Corning® Transwell® cell culture inserts (CLS3470), 4-phenylbutyric acid (4-PBA) (P21005) and FITC-Dextran (46945) were available from Sigma-Aldrich (St. Louis, MO, USA).

Cell culture

Bovine pulmonary artery endothelial cells (BPAEC) are available from Genlantis (San Diego, CA) and are maintained in DMEM (45000–304), in which we add fetal bovine serum and 1× penicillin/streptomycin. The cells are left to grow at 37°C, and in 5% CO2-95% air, and all materials used are obtained from VWR (Radnor, PA).Citation9

Western blot analysis

The process is based on the separation of proteins according to their molecular weight, which are consequently transferred to a nitrocellulose membrane, and incubated with the appropriate antibodies. To visualize the secondary antibody, we use the ChemiDocTM Touch Imaging System from Bio-Rad (Hercules, CA). Details on the procedures have been previously published.Citation9,Citation13,Citation14

Cell viability measurement

The cells were seeded onto 96-well culture plates in complete growth media and were treated with 4-PBA (0.1–20 mM) for 24 hours (H). The media was replaced with serum-free media containing 5 mg/mL MTT. An incubation of 3.5 H followed. DMSO dissolved the MTT crystals, and 0.25 H later absorbance (570 nm) was measured utilizing the SPECTROstar Nano® Absorbance Plate Reader by BMG LABTECH.

Fluorescein isothiocyanate (FITC)-Dextran permeability assay

BPAEC were treated with vehicle (0.1% DMSO) or 4-PBA (2 mM) prior to exposure to vehicle (PBS) or LPS (10 μg/ml) (2 H); and FITC-dextran was added. 20 minutes after, fluorescence intensity was measured utilizing a plate reader (see previous paragraph), as previously described.Citation9

Densitometry and statistical analysis

ImageJ software (National Institute of Health) was the tool to assess band density, and data are expressed as Mean ± SEM (standard error of the mean). Student’s t-test determined significance (P < 0.05). GraphPad Prism (version 5.01) provided statistical analysis, and n is the number of repeats.

Results

4-PBA suppresses BiP expression in BPAEC

Bovine lung cells were treated with vehicle (0.1% DMSO), or 4-PBA (2 mM) for 12 and 24 hours (H). The results demonstrate that 4-PBA significantly inhibits the expression of the UPR marker BiP ().

Figure 1. 4-PBA potentiates LPS-induced endothelial barrier dysfunction.

Western blot analysis of BiP and β-actin (a) after treatment of BPAEC with vehicle (0.1% DMSO) or 4-PBA (2 mM) for 12 hours (H) and 24 H. Western blot analysis of pSTAT3 and STAT3 (c), pERK1/2 and ERK1/2 (d) after treatment of BPAEC with vehicle (0.1% DMSO) or 4-PBA (2 mM) for 24 H and post-treatment with vehicle (PBS), or LPS (10 μg/ml) (2 H). The blots shown are representative of three independent experiments. The signal intensity of the bands was analyzed by densitometry. Protein levels of BiP, pSTAT3, and pERK1/2 were normalized to β-actin, STAT3, and ERK1/2, respectively. *P < 0.05 vs. vehicle (VEH) and $P < 0.05 vs. LPS. Mean ± SEM. (b) BPAEC were treated with either VEH (0.1% DMSO) or 4-PBA (0.1, 1, 2, 5, 10, 15, and 20 mM) for 24 H. Cell viability was evaluated by MTT assay. *P < 0.05 vs VEH, n = 4. Mean ± SEM. (e) BPAEC were seeded onto the trans-well inserts of a 24-well culture plate. After 24 H, the cells were pre-treated with VEH (0.1% DMSO) or 4-PBA (2 mM) for 24 H, and then exposed to VEH (PBS) or LPS (10 µg/ml) for 2 H. After that, cells were incubated with FITC-dextran (1 mg/ml). 20 minutes later, 100 μl of basal media was removed, and fluorescence intensity was measured. *P < 0.05 vs vehicle (VEH) and $P < 0.05 vs LPS. n = 4. Mean ± SEM.
Figure 1. 4-PBA potentiates LPS-induced endothelial barrier dysfunction.

Effects of 4-PBA on lung endothelial cell viability

The effects of 4-PBA on cell viability were measured utilizing MTT assay. Cells were seeded on a 96-well plate (10,000 cells/well) and were treated with either vehicle (0.1% DMSO) or 4-PBA (0.1–20 mM) for 24 hours (H). BPAEC treatment with 4-PBA at 0.1–5 mΜ did not significantly affect cell viability. However, higher doses (10–20 mM) of that UPR suppressor reduced cell viability ().

4-PBA potentiates LPS-induced STAT3 activation in BPAEC

The bovine cells were treated with vehicle (0.1% DMSO), or 4-PBA (2 mM) for 24 H, and were then exposed to vehicle (PBS) or LPS (10 µg/ml) for 2 H. The LPS-treated cells exerted higher expression of phosphorylated STAT3, compared to the vehicle-treated cells. Pre-treatment with 4-PBA potentiated LPS-induced STAT3 activation ().

Induction of pERK1/2 expression by 4-PBA and LPS in endothelial cells

The BPAEC were treated with vehicle (0.1% DMSO), or 4-PBA (2 mM) for 24 H prior to vehicle (PBS) or LPS (10 µg/ml) exposure (2 H). The cells treated with LPS or 4-PBA showed increased activation of ERK1/2, compared to the vehicle-treated cells. However, pre-treatment with 4-PBA did not significantly increase phospho (p) ERK1/2 expression, as compared to the LPS-treated cells ().

4-PBA enhances LPS-induced induction barrier dysfunction

FITC-dextran assay was used to evaluate paracellular permeability. BPAEC were seeded onto the trans-well inserts (125000 cells/insert) of a 24-well culture plate to form monolayer. Then, the cells were pretreated with vehicle (0.1% DMSO) or 4-PBA (2 mM) (24 H) prior to a 2-hour treatment with vehicle (PBS) or LPS (10 µg/ml). Our observations suggest that LPS increased paracellular permeability and 4-PBA potentiated that effect ().

Discussion

Inflammation has been associated with endothelial permeability, inducing the formation of endothelial gaps.Citation15 Those gaps can be transient (acute inflammation), or sustained (chronic inflammation). Tumor necrosis factor-α (TNF-α) induces permeability by activating the Ras homolog gene family, member A (RhoA)/Rho-associated protein kinase (ROCK) pathway to dissociate vascular endothelial protein tyrosine phosphatase (VE-PTP) from cadherin 5, type 2 or VE-cadherin, modulating tight junction proteins.Citation16 Inflammatory mediators such as nuclear factor-kappa B (NF-κB), extracellular signal-regulated kinase 1/2 (ERK1/2), and Janus kinase 2/signal transducer and activator of transcription 3 (JAK2/STAT3) also contribute in barrier dysregulation.Citation17 demonstrates that 4-PBA potentiates LPS-induced STAT3 activation, and shows that this UPR suppressor enhanced pERK1/2 phosphorylation; while it did not affect cell viability in moderate concentrations ().

Lung injury, both direct and indirect, inflicts severe respiratory disorders and death.Citation18 Medical countermeasures to suppress endothelial inflammation are of urgent need, both Heat shock protein 90 (Hsp90) inhibitorsCitation19 and Growth Hormone Releasing Hormone (GHRH) antagonistsCitation20,Citation21 may represent exciting therapeutic possibilities in lung inflammatory disease due to their anti-inflammatory activities.Citation8,Citation22 There is very little information on the role of UPR activation in barrier integrity. Recent evidence suggest that it is involved in the barrier-protective effects of growth hormone-releasing hormone antagonists (GHRHAnt)Citation17,Citation23 and heat shock protein 90 (Hsp90) inhibitorsCitation24 in the vasculature.Citation25

GHRH regulates GH secretion, but it has also been involved in a variety of physiological processesCitation26. Receptors responding to that hormone are expressed in extra-hypothalamic tissues and endothelial cells,Citation27 including those of the brain barrier.Citation28 GHRH receptors are involved in inflammation, and GHRHAnt exert strong anti-inflammatory and anti-oxidative effects.Citation27,Citation29,Citation30 Those antagonists can inhibit bleomycin-induced fibrosisCitation31 and prevent LPS-induced bronchoalveolar lavage fluid (BALF) protein concentration in vivo, suggesting their protective effects against edema.Citation32 Hsp90 inhibitors exert similar activities, they can activate UPR,Citation23,Citation24 and counteract the kifunensine (UPR suppressor)-induced lung endothelial dysfunction.Citation23,Citation33 They act via the suppression of activated Hsp90, a molecular chaperone in charge with protein stability.Citation34 Interestingly, global UPR modulation due to Brefeldin A and Kifunensine affected endothelial cytoskeletal remodeling by cofilin and myosin light chain 2 (MLC2) activation regulation.Citation35,Citation36 Cofilin is phosphorylated by Rac1 and severs actin filaments, whereas MLC2 activation signals filamentous actin formation.Citation9,Citation17 Hence, the Rac1/pCofilin axis strengthens barrier function, and RhoA/MLC2 activation induces hyperpermeability responses.Citation37 P53 has been shown to mediate the aforementioned activities,Citation38,Citation39 and suppresses STAT3.Citation40

Endothelial function can be assessed in vitro via measurements of paracellular and transendothelial permeability. suggests that LPS increases paracellular permeability, and that 4-PBA augments LPS-induced vascular leakage as measured with Dextran-FITC. Indeed, this UPR suppressor itself impaired the endothelium barrier, since the bovine pulmonary artery endothelial cells treated with hat compound presented with barrier dysfunction.

ATF6-related studies revealed that this UPR sensor can support the endothelial barrier integrity.Citation23,Citation36 The ATF6 inducer AA147 counteracted LPS-induced endothelial barrier disruption by suppressing cofilin and MLC2 activation, as well as by reducing VE-cadherin phosphorylation. ATF6 suppression exerted the opposite effectsCitation13. Furthermore, PERK-knockout mice presented fibrosis,Citation41 and BiP silencing suppressed NF-κB activation, and augmented inflammation.Citation18,Citation42

In conclusion, our study suggest that 4-PBA potentiates LPS-triggered pSTAT3 activation, as well as LPS-induced increase in paracellular permeability. Moreover, UPR suppression in BPAEC resulted to ERK1/2 activation, and this UPR suppressor did not affect cell viability at moderate concentrations. The molecular events beyond those events are to be elucidated in future studies.

Disclosure statement

No potential conflict of interest was reported by the authors.

Additional information

Funding

Our work is supported by the Institutional Development Award (IDeA) from NIGMS/NIH (3P20GM103424-21).

References

  • Claesson-Welsh L, Dejana E, McDonald DM. Permeability of the endothelial barrier: identifying and reconciling controversies. Trends Mol Med. 2021;27(4):1–6. doi:10.1016/j.molmed.2020.11.006.
  • Sukriti S, Tauseef M, Yazbeck P, Mehta D. Mechanisms regulating endothelial permeability. Pulm Circ. 2014;4(4):535–551. doi:10.1086/677356.
  • Komarova Y, Malik AB. Regulation of endothelial permeability via paracellular and transcellular transport pathways. Annu Rev Physiol. 2010;72(1):463–493. doi:10.1146/annurev-physiol-021909-135833.
  • Barabutis N. Unfolded protein response in the COVID-19 context. Aging Health Res. 2021;1(1):100001. doi:10.1016/j.ahr.2020.100001.
  • Akhter MS, Uddin MA, Kubra K-T, Barabutis N. Autophagy, unfolded protein response and lung disease. Curr Res Cell Biol. 2020;1:1. doi:10.1016/j.crcbio.2020.100003.
  • Barabutis N. Unfolded protein response supports endothelial barrier function. Biochimie. 2019;165:206–209. doi:10.1016/j.biochi.2019.08.007.
  • Barabutis N. Unfolded protein response in acute respiratory distress syndrome. Lung. 2019;197(6):827–828. doi:10.1007/s00408-019-00279-4.
  • Barabutis N. Unfolded protein response: a regulator of the endothelial barrier. Endocr Metab Sci. 2021;3:3. doi:10.1016/j.endmts.2021.100092.
  • Kubra KT, Uddin MA, Barabutis N. Tunicamycin protects against LPS-Induced lung injury. Pharmaceuticals (Basel). 2022;15(2):134. doi:10.3390/ph15020134.
  • Xia T, Liao Y-Q, Li L, Sun L-Y, Ding N-S, Wu Y-L, Lu K-L. 4-PBA attenuates fat accumulation in cultured spotted seabass fed high-fat-diet via regulating endoplasmic reticulum stress. Metabolites. 2022;12(12):1197. doi:10.3390/metabo12121197.
  • Dong Y, Li L, Xia T, Wang L, Xiao L, Ding N, Wu Y, Lu K. Oxidative stress can be attenuated by 4-PBA caused by high-fat or ammonia nitrogen in cultured spotted seabass: the mechanism is related to endoplasmic reticulum stress. Antioxid (Basel). 2022;11(7):1276. doi:10.3390/antiox11071276.
  • Rubenstein RC, Zeitlin PL. Sodium 4-phenylbutyrate downregulates Hsc70: implications for intracellular trafficking of DeltaF508-CFTR. Am J Physiol Cell Physiol. 2000;278(2):C259–67. doi:10.1152/ajpcell.2000.278.2.C259.
  • Kubra KT, Akhter MS, Saini Y, Kousoulas KG, Barabutis N. Activating transcription factor 6 protects against endothelial barrier dysfunction. Cell Signal. 2022;99:110432. doi:10.1016/j.cellsig.2022.110432.
  • Kubra KT, Barabutis N. P53 in endothelial function and unfolded protein response regulation. Cell Biol Int. 2022;46(12):2257–2261. doi:10.1002/cbin.11891.
  • Barabutis N, Verin A, Catravas JD. Regulation of pulmonary endothelial barrier function by kinases. Am J Physiol Lung Cell Mol Physiol. 2016;311(5):L832–L845. doi:10.1152/ajplung.00233.2016.
  • Fang M, Zhong W-H, Song W-L, Deng Y-Y, Yang D-M, Xiong B, Zeng H-K, Wang H-D. Ulinastatin ameliorates pulmonary capillary endothelial permeability induced by sepsis through protection of tight junctions via inhibition of TNF-α and related pathways. Front Pharmacol. 2018;9:823. doi:10.3389/fphar.2018.00823.
  • Barabutis N, Akhter MS, Kubra KT, Jackson K. Growth Hormone-Releasing Hormone in Endothelial Inflammation. Endocrinology. 2022;164(2):bqac209. doi:10.1210/endocr/bqac209.
  • Barabutis N. Unfolded protein response in lung health and disease. Front Med. 2020;7:344. doi:10.3389/fmed.2020.00344.
  • Barabutis N. Heat shock protein 90 inhibition in the inflamed lungs. Cell Stress Chaperones. 2020;25(2):195–197. doi:10.1007/s12192-020-01069-1.
  • Barabutis N, Akhter MS, Kubra K-T, Jackson K. Growth hormone–releasing hormone in endothelial inflammation. Endocrinology. 2022;164(2). doi:10.1210/endocr/bqac209.
  • Barabutis N, Kubra KT, Akhter MS. Growth hormone-releasing hormone antagonists protect against hydrochloric acid-induced endothelial injury in vitro. Environ Toxicol Pharmacol. 2023;99:104113. doi:10.1016/j.etap.2023.104113.
  • Akhter MS, Uddin MA, Kubra K-T, Barabutis N. Elucidation of the molecular pathways involved in the protective effects of AUY-922 in LPS-Induced inflammation in mouse lungs. Pharmaceuticals (Basel). 2021;14(6):522. doi:10.3390/ph14060522.
  • Akhter MS, Uddin MA, Schally AV, Kubra K-T, Barabutis N. Involvement of the unfolded protein response in the protective effects of growth hormone releasing hormone antagonists in the lungs. J Cell Commun Signal. 2021;15(1):125–129. doi:10.1007/s12079-020-00593-0.
  • 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;6:6. doi:10.1016/j.medidd.2020.100046.
  • Barabutis N. Unfolded protein response in endothelial injury. Cell Cycle. 2022;21(19):2009–2012. doi:10.1080/15384101.2022.2082024.
  • Barabutis N, Schally AV. Growth hormone-releasing hormone: extrapituitary effects in physiology and pathology. Cell Cycle. 2010;9(20):4110–4116. doi:10.4161/cc.9.20.13787.
  • Uddin MA, Akhter MS, Singh SS, Kubra K-T, Schally AV, Jois S, Barabutis N. GHRH antagonists support lung endothelial barrier function. Tissue Barriers. 2019;7(4):1669989. doi:10.1080/21688370.2019.1669989.
  • Barabutis N, Akhter MS, Uddin MA, Kubra K-T, Schally AV. GHRH antagonists protect against hydrogen peroxide-induced breakdown of brain microvascular endothelium integrity. Horm Metab Res. 2020;52(5):336–339. doi:10.1055/a-1149-9347.
  • Kubra KT, Akhter MS, Apperley K, Barabutis N. Growth hormone-releasing hormone antagonist JV-1-36 suppresses reactive oxygen species generation in A549 lung cancer cells. Endocrines. 2022;3(4):813–820. doi:10.3390/endocrines3040067.
  • Du L, Ho BM, Zhou L, Yip YWY, He JN, Wei Y, Tham CC, Chan SO, Schally AV, Pang CP, et al. Growth hormone releasing hormone signaling promotes Th17 cell differentiation and autoimmune inflammation. Nat Commun. 2023;14(1):3298. doi:10.1038/s41467-023-39023-1.
  • Zhang C, Cai R, Lazerson A, Delcroix G, Wangpaichitr M, Mirsaeidi M, Griswold AJ, Schally AV, Jackson RM. Growth hormone-releasing hormone receptor antagonist modulates lung inflammation and fibrosis due to bleomycin. Lung. 2019;197(5):541–549. doi:10.1007/s00408-019-00257-w.
  • Akhter MS, Kubra K-T, Uddin MA, Jois S, Barabutis N. An antagonist of growth hormone-releasing hormone protects against LPS-induced increase of bronchoalveolar lavage fluid protein concentration. Inflamm Res. 2022;71(2):183–185. doi:10.1007/s00011-021-01531-5.
  • Kubra KT, Uddin MA, Akhter MS, Barabutis N. Luminespib counteracts the Kifunensine-induced lung endothelial barrier dysfunction. Curr Res Toxicol. 2020;1:111–115. doi:10.1016/j.crtox.2020.09.003.
  • Maehana T, Tanaka T, Hashimoto K, Kobayashi K, Kitamura H, Masumori N. Heat shock protein 90 is a new potential target of anti-rejection therapy in allotransplantation. Cell Stress Chaperones. 2022;27(4):337–351. doi:10.1007/s12192-022-01272-2.
  • 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(2):C214–C220. doi:10.1152/ajpcell.00142.2021.
  • 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.
  • Barabutis N. P53 in RhoA regulation. Cytoskeleton (Hoboken). 2020;77(5–6):197–201. doi:10.1002/cm.21604.
  • 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(8):L776–87. doi:10.1152/ajplung.00334.2014.
  • 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(3):1792–1804. doi:10.1111/jcmm.13460.
  • Pham TH, Park H-M, Kim J, Hong JT, Yoon D-Y. STAT3 and p53: dual Target for Cancer Therapy. Biomedicines. 2020;8(12):637. doi:10.3390/biomedicines8120637.
  • Liu X, Kwak D, Lu Z, Xu X, Fassett J, Wang H, Wei Y, Cavener DR, Hu X, Hall J, et al. Endoplasmic reticulum stress sensor protein kinase R–Like endoplasmic reticulum kinase (PERK) protects against pressure overload–induced heart failure and lung remodeling. Hypertension. 2014;64(4):738–744. doi:10.1161/HYPERTENSIONAHA.114.03811.
  • Leonard A, Paton AW, El-Quadi M, Paton JC, Fazal F. Preconditioning with endoplasmic reticulum stress ameliorates endothelial cell inflammation. PLoS One. 2014;9(10):e110949. doi:10.1371/journal.pone.0110949.