Abstract
Cells respond to stress by activating a variety of defense signaling pathways, including cell survival and cell death pathways. While cell survival signaling helps the cell to recover from acute insults, cell death or senescence pathways induced by chronic insults can lead to unresolved pathologies. Arterial hypertension results from chronic physiological maladaptation against various stressors represented by abnormal circulating or local neurohormonal factors, mechanical stress, intracellular accumulation of toxic molecules and dysfunctional organelles. Hypertension and aging share common mechanisms that mediate or prolong chronic cell stress, such as endoplasmic reticulum stress and accumulation of protein aggregates, oxidative stress, metabolic mitochondrial stress, DNA damage, stress-induced senescence and pro-inflammatory processes. This review discusses common adaptive signaling mechanisms against these stresses including unfolded protein responses, antioxidant response element signaling, autophagy, mitophagy and mitochondrial fission/fusion, signaling effector stimulator of interferon genes (STING)-mediated responses and activation of pattern recognition receptors. The main molecular mechanisms by which the vasculature copes with hypertensive and aging stressors are presented and recent advancements in stress-adaptive signaling mechanisms as well as potential therapeutic targets are discussed.
Keywords: vascular senescence, endoplasmic reticulum stress, mitochondria, inflammation
Introduction
Cells respond to stress by modulating many signaling pathways ranging from cell survival to cell death pathways. Initial responses to a stressful stimulus normally aid in cell recovery from insult to maintain organismal homeostasis1–3. However, if the stress stimulus persists and/or the cell has a poor adaptive capacity to resolve the insult, the stress responses become stronger, chronic and lead to a secondary (and usually one-way) wave of stress-signaling pathways, including death pathways (apoptosis, necroptosis, pyroptosis) or senescence pathways2.
The scenario is more complex if we consider that i) multiple stress-response pathways can be simultaneously activated or recruited at different time points, ii) there is an interplay among signaling pathways, and iii) cell signaling is influenced across tissues, organs or the whole organism by physiological and pathological processes, such as aging and development of cardiovascular diseases (CVD) including hypertension, which is the topic of this compendium.
Accordingly, it has become a consensus that the development of hypertension is a result of chronic physiological maladaptation against various stressors. These include neurohormonal factors [norepinephrine/epinephrine, angiotensin II (Ang II), aldosterone, anti-diuretic hormone or vasopressin], circulating or local bioactive substances derived from immune cells, adipose tissue, skeletal muscle and endothelial cells, mechanical stresses (arterial pressure and shear stress), intracellular accumulation of metabolites (carbon dioxide, hydrogen and potassium ions) and toxic molecules as well as dysfunctional organelles4–7. In the development of hypertension, chronic maladaptation against these insults eventually leads to disruption of cellular homeostasis, likely contributing to worsened blood pressure control and hypertensive complications. Sedentary lifestyle, excess caloric intake, obesity, hyperglycemia and aging likely exaggerate these processes. It is therefore very important to better understand the molecular mechanisms by which affected organs, such as the vasculature, cope with “hypertensive” stressors at cellular and organelle levels. Taking this opportunity, we will discuss recent advancements in stress-adaptive signaling mechanisms as potential therapeutic targets for people with hypertension and pre-hypertensive conditions such as those with early vascular aging.
Animal Models and Limitations
Animal models of primary hypertension have been developed and extensively reviewed elsewhere8. The main experimental models include Ang II infusion, deoxycorticosterone acetate (DOCA)-salt, spontaneously hypertensive rats (SHR), and nitric oxide (NO) synthase inhibition by L-N-nitro arginine methyl ester (L-NAME). The most cited model, chronic Ang II infusion mimics the activation of the renin-angiotensin aldosterone system (RAAS) and generates most of the end-organ injuries seen in human hypertension9. Although studies with Ang II infusion have significantly contributed to our understanding of hypertension, the inability to mimic aging and other genetic or environmental stressors seen in human hypertension is a limitation. In rodent models, “adult age” is usually between 6 and 20 weeks, likely contributing to the lack of translation into clinical outcomes9. To overcome these limitations, more rigorous studies have begun to combine multiple models such as aging, salt-sensitive hypertension, and metabolic dysfunction in rodents10, 11. In the clinical practice, RAAS blockade, regardless of mono- or combination therapy, does not show the desired level of end-organ protection or reduction in mortality, although it effectively reduces blood pressure12, 13. This indicates that several non-RAAS factors contribute to hypertensive end-organ damage. Obviously, the use of Ang II model alone may lead to incomplete conclusions on the impact of a given stress signaling in hypertension. While most studies cited in this review use a single (Ang II) model, with a few studies using two distinct models, it is important to reconcile the stress mechanisms discussed here with multiple hypertension models for better knowledge as well as for potential translation.
Premature Vascular Aging Associated with Common Stress Signaling
In a disease-free middle-aged population, there is a subset of people whose vasculature demonstrates a significant sign of premature aging, i.e. endothelial dysfunction and arterial stiffness. They are thus considered a high-risk population for future development of hypertension and coronary artery disease. This condition, also known as early vascular aging (EVA) syndrome, is frequently associated with hypertension14, 15. In contrast, there is another chronologically middle-aged population whose vasculature is as healthy as much younger populations [Healthy or Super Normal Vascular Aging (HVA/SUPERNOVA)]15. This group of people is hypothesized to be protected from CVD as well as other aging-accelerated disorders, such as neurodegenerative diseases, including Alzheimer’s16. This perspective represents a growing field of study in which ‘biological age’, rather than chronological age, can be therapeutically targeted. Although the molecular mechanisms involved in the development of EVA in humans remain unclear, environmental exposure to several chronic stressors (over nutrition, sedentary lifestyle, smoking, alcohol e.g.) seem to be involved15. Accordingly, this review article addresses the intracellular stress-sensing and -responding signaling mechanisms that are likely relevant for hypertension development and progression and with potential for add-on therapies.
Both aging (regardless of chronological or biological/EVA) and arterial hypertension are linked to functional, structural and mechanical vascular changes. Characteristic vascular abnormalities in aging and hypertension include endothelial dysfunction, calcification and increased stiffness, remodeling and chronic low-grade inflammation17. Common mechanisms involved in vascular dysfunction in hypertension and other aging-related CVD likely include (but are not limited to) those that mediate or prolong chronic cell stress, such as 1. endoplasmic reticulum (ER) stress and accumulation of protein aggregates, 2. oxidative stress, 3. metabolic stress centered at mitochondria, 4. DNA damage, 5. stress-induced senescence and senescence-associated secretory phenotype (SASP), and 6. generation of pro-inflammatory factors and processes by danger signals. There also are common adaptive mechanisms against these stresses including unfolded protein responses (UPR), antioxidant response element (ARE) signaling, autophagy, mitophagy and mitochondrial fusion, induction of signaling effector stimulator of interferon genes (STING) pathway and senolysis (elimination of senescent cells).
ER Stress Responses in Hypertension
Hypertensive animal models confirm the presence of aortic ER stress markers and pharmacological inhibition via chemical chaperone 4-phenylbutyric acid (4-PBA) and/or tauroursodeoxycholic acid (TUDCA) reduces fibrosis, inflammation, vascular hypercontractility, and increased blood pressure18–21. Single cell transcriptomics of hearts from angiotensin II (Ang II)-infused mice identified ER stress elements to be highly involved in hypertensive cardiac remodeling22. Other studies found that Ang II induces protein aggregate formation in the heart23 and kidney24. At the cellular level, a variety of extracellular stressors and humoral factors, including Ang II, elicit induction of ER stress/UPR markers in endothelial cells (ECs), vascular smooth muscle cells (VSMCs), cardiac myocytes and cardiac fibroblasts25. Moreover, induction of protein aggregate accumulation and UPR were seen in VSMCs stimulated with Ang II26, suggesting that Ang II-induced proteotoxicity in the vasculature is a novel target for hypertension therapy. Enhanced ER stress/UPR within biopsied endothelial cells has been found in obese adults with endothelial dysfunction27. An ongoing clinical trial is testing TUDCA effects on vascular dysfunction in older and obese individuals (Clinicaltrials.gov identifier: NCT04001647). However, how ER dynamics and protein aggregates contribute to hypertension and vascular damage require further investigation.
UPR and ER-Associated Degradation
The ER is an essential organelle for protein synthesis and calcium (Ca2+) handling. Chaperones such as HSP47 and glucose-regulated protein 78 (GRP78) aid in properly folding proteins into tertiary structures28. When imbalances in protein synthesis, reduction-oxidation (redox) status, or Ca2+ handling occur, three transmembrane ER sensors initiate the stress-induced UPR to maintain homeostasis (proteostasis). These include activating transcription factor 6 (ATF6), double-stranded RNA-activated protein kinase-like ER kinase (PERK), and inositol-requiring transmembrane kinase 1 alpha (IRE1α). Transcription factors induced by UPR activation bind ER stress response elements, leading to the expression of chaperones, ER-associated degradation (ERAD) genes, and autophagy machinery, thus restoring ER protein folding competency29.
Under chronic ER stress however, UPR signaling seems maladapted and likely contributes to hypertension pathology. C/EBP homologous protein (CHOP) activation of caspases and BAX/BIM release of cytochrome C leads to apoptosis30. Ang II infusion increases CHOP and ATF6 in vivo, and global CHOP deficient mice are protected from hypertension-mediated cardiac hypertrophy, fibrosis and vascular dysfunction31. In VSMC from SHR, compared with those from WKY, NAPDH oxidases are increasingly localized in the ER, which overall contributes to increased oxidative stress. Moreover, treatment with 4-PBA reduces vascular dysfunction in stroke-prone SHR (SHRSP)18. In a combination model of hypertensive chronic kidney disease (CKD) utilizing Ang II infusion plus DOCA-salt, 4-PBA treatment not only reduced blood pressure and renal ER stress markers, but significantly attenuated gene expression of inflammatory and fibrotic markers32. These observations suggest that while the UPR is considered a defense mechanism against proteotoxicity, over-activation of UPR is detrimental and contributes to hypertension.
Pharmacological targeting of the individual ER UPR arms, instead of general ER stress, via 4-PBA and TUDCA have yielded promising results. The ATPase vasolin-containing protein (VCP) is involved in ERAD signaling and is reduced in SHR. Overexpression of VCP attenuates Ang II-induced hypertrophy in vitro via mTOR signaling, and reduces cardiac dysfunction associated with pressure overload in mice33. Overall, VCP activity enhancement to reduce hypertrophy seems to protect against hypertensive remodeling. However, its role in vascular cells remains undetermined. In high-salt diet hypertensive mouse models, vascular expression of the ER stress-dependent transcription factor ATF4 is increased and regulates blood pressure and endothelial function. Furthermore, gut-microbiota composition in salt-sensitive hypertensive mice can be targeted via potassium supplementation to reduce ATF4 expression and, subsequently, blood pressure34. Pharmacological inhibition of IRE1α-XBP1 branch has anti-fibrotic and anti-inflammatory effects via an IRE1α inhibitor, KIRA6 or STF-08310 35–37. Overall, small molecule inhibitors targeting each arm of UPR, including the IRE1α UPRsome branch as well as ERAD machinery, are promising opportunities to reverse hypertension and its vascular consequences.
To identify upstream therapeutic targets to reduce hypertensive ER stress, protein aggregation has been explored. Adenoviral GRP78 delivery attenuated Ang II-induced protein aggregate formation and inflammatory monocyte adhesion to VSMCs28. Moreover, pre-amyloid oligomers were formed in VSMCs in response to Ang II stimulation, which were attenuated with 4-PBA treatment26. In hypertension-associated atrial fibrillation, lipid oxidation develops and contributes to amyloid deposition in the heart. Scavenging these reactive peroxidation lipid products via chaperone 3-HOBA reduces atrial amyloid burden and blood pressure in Ang II-infused mice38. These observations suggest a potential application of protein chaperoning therapy against hypertension and related CVD in consonance with clinical trials for age-associated neurodegenerative diseases.
Effects of ER stress inhibition in hypertension have been summarized to allow an overall understanding of therapeutic targets in the vasculature (Figure 1, Table 1). The UPR pathway can be either adaptive or chronic and potentially maladaptive. The adaptive arm has developed to maintain ER function and proteostasis, whereas the chronic arm sustains inflammation, causes organ damage, induces senescence, or eliminates dysfunctional cells via apoptosis. Such maladaptive ER UPR occurs in hypertension due to increased protein synthesis demand via hypertrophic stimuli39, 40 and oxidative stress18. Therefore, restoration of normal proteostasis can protect the vasculature and organs from damage in addition to RAAS inhibition.
Figure 1. Pharmacological targets of ER stress signaling in hypertension.

AT1R signaling disrupts ER homeostasis via increased protein synthesis demand and oxidative stress. Adaptive stress responses including the UPR, ER mediated autophagy (ER-phagy), and ER associated degradation are induced. These responses are initiated by chaperone GRP78 detachment from three transmembrane molecules in order to aid in refolding of misfolded proteins. IRE1α oligomerizes and autophosphorylates to induce RNAse activity to alternatively splice XBP1 mRNA to XBP1s. XBP1s serves as a transcription factor binding to ER stress response elements (ERSE) to induce transcription of cytokines, lipid biogenesis, chaperones, and ERAD. IRE1α also acts as a scaffold for TRAF2 docking and initiation of proinflammatory JNK and NF-κB signaling. PERK dimerizes and autophosphorylates leading to phosphorylation of eIF2α and translation attenuation in an attempt to reduce overwhelming demand of protein folding on the ER. There is an increase translation in ATF4 expression which acts as a transcription factor for amino acid metabolism, autophagy elements, and an antioxidant response. However, CHOP expression is also induced which leads to apoptosis. GRP78 detachment from ATF6 results in its translocation to the Golgi where it is cleaved by S1 and S2 proteases resulting in ATF6 cleaved for which transcribes chaperones, ERAD elements, and XBP1. ERAD induction is mediated by VCP/p97, a Ca2+-associated ATPase which participates in ubiquitin-proteasome system to degraded misfolded proteins by interacting with E3 ubiquitin ligases such as HRD1. ER-phagy involves PI3K complexes which form at ER tubules to recruit autophagic initiation complexes, leading to the lipidation of LC3 and formation of autophagosome. Created with BioRender.com
Table 1.
Potential targets in stress-elicited signaling pathways
| Potential Targets | Therapeutic approach | Outcome | Reference |
|---|---|---|---|
| Endoplasmic Reticulum | inhibition via 4-PBA or TUDCA) | reduces fibrosis, inflammation, vascular hypercontractility, BP | 18–21 |
| attenuates Ang II-induced formation of pre-amyloid oligomers | 26 | ||
| prevents Ang II-induced senescence in VSMCs and ECs | 26, 92 | ||
| Unfolded Protein Responses | GRP78 overexpression | protects against I/R injury | 230 |
| adenoviral GRP78 delivery | attenuates Ang II-induced protein aggregate formation and monocyte adhesion to VSMCs | 28 | |
| compound 147 (cleaves ATF6) | protects against cardiomyocyte oxidative stress | 231 | |
| IRE1α- XBP1 inhibition | anti-fibrotic and anti-inflammatory effects | 35–37 | |
| CHOP knockdown | attenuates cardiac hypertrophy, fibrosis and altered vascular reactivity | 31 | |
| Mitochondrial Stress | SIRT3 overexpression | improves endothelial function and attenuates Ang II-induced hypertension | 49 |
| SIRT3/SOD2 activation | increases endothelial reparative capacity of progenitor cells | 50 | |
| chaperone 3-HOBA | decreases vascular dysfunction and hypertension | 51 | |
| FAM3A knockdown | reduces Ang II-mediated cardiac and vascular remodeling, vasoconstriction, and hypertension | 52 | |
| Mitochondrial Fission/Fusion and Mitophagy | 17-DMAG (inhibits HSP90) | inhibits Ang II-induced mitochondrial fission and adventitial remodeling | 59 |
| DRP1 (inhibition or knockdown) | protects against Ang II-induced AAA | 60 | |
| cGAS-STING | STING (inhibition or knockdown) | reduces ATAAD incidence in mice prevents Ang II-induced abnormal vascular reactivity reduces pro-apoptotic, pro-necroptotic, pro-pyroptosis, and pro-inflammatory signaling prevents EC inflammation | 71 |
| NRF2 | tBHQ (activator of Nrf2) | prevents Ang II-induced endothelial dysfunction, remodeling, and hypertension | 116 |
| Nrf2 deletion | increases sympathetic outflow, impairs baroreflex function and induces hypertension | 121 | |
| bardoxolone/L-sulforaphane (NRF2 activators) | improve vascular function and reduces Ang II-induced ROS generation | 117 | |
| TLR | TLR2 knockdown | prevents impaired vascular tone, iNOS-derived NO formation and TNF-α production prevents Ang II-induced aortic fibrosis and endothelial to mesenchymal transition | 158,159 |
| VIPER (TLR4 blocker) | prevents hypertension induced by abnormal HDL containing symmetric dimethylarginase reduces BP, HMGB1, and TNF-α, IL-1β expression | 168,170 | |
| TAK242 (TLR4 inhibitor) | reduces BP and expression of TLR4, MyD88, NF-κB, TNF-α and IL-1β | 169 | |
| anti-TLR4 | decreases BP and reduces expression of TLR4, COX-2, TxA2, IL-6 | 171 | |
| TLR4 silencing | attenuates Ang II-induced cardiac hypertrophy | 174 | |
| TLR3 silencing | prevents Ang II-induced increased BP and cardiac hypertrophy | 174 | |
| NLPR3 | Canakinumab (human monoclonal IL-1β antibody) | reduces cardiovascular events and hospitalization decreases hypertension and renal fibrosis | 211–213, 232 |
| anakinra (IL-1R antagonist) | prevents hypertension-associated cardiovascular dysfunction and oxidative stress. attenuates hypertension, vascular remodeling, | 214 | |
| NLRP3 (gene silencing or blockade) | reduces aldosterone-induced vascular damage | 193 | |
| MCC950 (NLRP3 inhibitor) | reduces BP and renal damage and dysfunction in salt-sensitive hypertension | 209 | |
| restores EC function and reduces stress oxidative and inflammatory processes | 192 | ||
| reduces cardiac damage and improves insulin and leptin sensitivity | 224, 225, 228 |
4-PBA, 4-Phenylbutyric acid; AAA, aortic abdominal aneurism; Ang II, angiotensin II; BP, blood pressure; ECs, endothelial cells; FAM3A (family with sequence similarity 3 member A);heat shock protein 90 (HSP90); I/R, ischemia/reperfusion; iNOS, inducible nitric oxide synthase; tBHQ, tert-butylhydroquinone; TUDCA, tauroursodeoxycholic acid; VSMCs, vascular smooth muscle cells.
Autophagy and ER Stress in Hypertension
While autophagic responses are involved in a wide variety of homeostatic and adaptative adjustments against various stresses conditions, autophagy also plays a key role in alteration of ER stress by removing toxic protein aggregates termed aggrephagy. In several CVD models, including Ang II-induced hypertension and cardiovascular remodeling, both pharmacological and genetic manipulation of autophagy have been conducted.
For example, Ang II was infused in heterozygous mice for the autophagy protein 5 (Atg5), an E3 ubiquitin ligase that mediates autophagosome formation. In control wild type mice, Ang II infusion enhanced autophagy, assessed by induction of Atg5, and increased LC3-II/LC3-I ratio. Mitophagy in macrophages was also enhanced in Ang II-infused mice. As expected for the homeostatic roles of autophagy and mitophagy, Ang II-induced cardiac fibrosis was exaggerated in Atg5+/− mice and this was associated with enhanced reactive oxygen species (ROS) production and NF-κB activation in macrophages41. In line with these observations, in hearts of a swine model of renovascular hypertension, Ang II type 1 receptor (AT1R) antagonism attenuated markers of enhanced autophagy and mitophagy42. However, it remains unclear whether Ang II signaling or hypertension drives autophagy in mice, while Ang II-induced autophagy is observed in VSMCs43. In SHR, supplementation with the autophagy inducer trehalose did not change blood pressure. However, mesenteric resistance arteries displayed preserved endothelial function and reduced arterial stiffness44. Under chronic ER stress, ATF4-induced autophagy is known to function at overwhelming ERAD capacity45 and this is a part of the common integrated stress response pathways46. It is therefore necessary to further evaluate the relationship between UPR and autophagy in hypertension.
Mitochondrial Stress Response in Hypertension
Mitochondria are the powerhouse of cellular and energy metabolism. The mitochondrial outer membrane directly interacts with the ER membrane and regulates fission/fusion events, whereas the inner mitochondrial membrane is particularly involved in oxidative phosphorylation, ATP generation, Ca2+ signaling, and protein import. These dynamic functions play critical roles in cell metabolism, antioxidant capacity, inflammation, growth, proliferation, and apoptosis. Accordingly, mitochondrial dysfunction has been identified in both human and animal models of hypertension47, however investigations to identify pharmacological targets of intervention are still ongoing.
Mitochondrial Oxidative Stress and the Defense System
Metabolic disorders and oxidative stress contribute to the pathogenesis of vascular dysfunction, including hypertension48. During normal cellular respiration, utilization of the mitochondrial electron transport chain (ETC) to synthesize large quantities of ATP36 requires a series of univalent reductions, leading sequentially to the production of superoxide anion (O2.−).
Antioxidant enzymes have evolved to limit oxidized byproducts from interrupting normal cellular physiology. One such class of enzymes, sirtuins (SIRT) are critical in the redox conversion of O2.− to hydrogen peroxide (H2O2). Arterioles from hypertensive humans have significantly reduced mitochondrial SIRT3, and global SIRT3 depletion enhances vascular O2.−, blood pressure, hypertrophy, permeability, senescence, and inflammation in Ang II and DOCA-salt mouse models of hypertension49. Thus, transgenic SIRT3 overexpression improves endothelial function and attenuates Ang II-induced hypertension, suggesting that this mitochondrial target has therapeutic potential.
Superoxide dismutases (SODs) are part of a major antioxidant defense system, and manganese SOD2 in the mitochondria is regulated by deacetylation via SIRT3 activity. Endothelial progenitor cells from hypertensive patients have impaired reparative capacity, which can be mitigated by SIRT3/SOD2 signaling50. Reactive dicarbonyl lipid peroxidation products, isolevuglandins are increased in hypertensive patients, and mitochondria-targeted scavenging of these oxidative stress byproducts using chaperone 3-HOBA has therapeutic potential to treat vascular dysfunction and hypertension51. A novel mitochondrial protein, FAM3A (family with sequence similarity 3 member A), enhances ATP production and is increased in arteries of hypertensive mice. VSMCs specific FAM3A deletion reduces Ang II-mediated cardiac and vascular remodeling, vasoconstriction, and increased blood pressure in mice52. While these observations, summarized at Table 1, support therapeutic potential in manipulating mitochondrial oxidative stress or energy production in hypertension, clinical trials appear to have mixed outcomes.
Interestingly, both Ang II AT1R and AT2R are localized within the mitochondria53, and intracellular Ang II fusion protein selectively expressed in the mitochondria of renal proximal tubules cells regulates ETC complexes, inhibits proximal tubules sodium reabsorption, natriuretic response and contributes to hypertension54. These findings have not been explored in other cell types and may represent a new area of hypertension research.
Mitochondrial Fission/Fusion and Mitophagy
Although ATP generation is the primary function of mitochondria, mitochondrial dynamics affect the non-bioenergetic capacity of cells. Fission and fusion of mitochondria control energy homeostasis within the cell while imbalances may contribute to CVD such as obesity, heart failure, and hypertension55. The homeostatic balance can lean towards fission to create smaller mitochondria, whereas fusion results in a more interconnected mitochondrial network. Both processes are regulated by guanosine triphosphatases (GTPases) and non-GTPase receptor proteins. For fission, dynamin-related protein 1 (DRP1) is targeted to the outer mitochondrial membrane by mitochondrial fission factor56. Excessive fission is a signal of increased mitochondrial damage and leads to enhanced mitophagy, i.e. clearance of damaged mitochondria via autophagosome engulfment and lysosomal degradation57. Fusion is mediated by mitofusins 1 and 2 (MFN1 and MFN2) in the outer membrane, and optic atrophy 1 (OPA1) in the inner membrane58.
Hypertensive stimuli augment the vascular cell expression of mitochondrial dynamic proteins. In adventitial fibroblasts, Ang II treatment increases mitochondria fission, collagen and α-SMA which can be inhibited with DRP1 silencing59. Moreover, pharmacological inhibition of heat shock protein 90 (HSP90) with 17-DMAG blocks Ang II-induced calcineurin regulation of DRP-1-mediated mitochondrial fission and adventitial remodeling59. In human abdominal aortic aneurysm (AAA) samples, DRP1 is increased in the aortic medial layer, and pharmacological inhibition of DRP1 by mdivi1, or heterozygous knockdown protects against Ang II-induced AAA in mice60. While phosphorylation sites primarily act to induce or inhibit DRP1 oligomerization around mitochondria, a high fat diet has been shown to increase DRP1 acetylation in the heart and to contribute to cardiomyocyte dysfunction and death61. In ECs, DRP1 activation enhances inflammation and monocyte recruitment62, and in macrophages, it promotes intimal thickening after vascular injury63. Moreover, in SHR,4- week mdivi1 treatment reduced hypertension and medial aortic hypertrophy, which were associated with reduction of inflammatory cytokines64. Whereas evidence supports that mitochondria fission mediates vascular hypertensive responses, limited studies have explored fusion protein regulation in this process.
Mitophagy is inherently tied to autophagy activation, which has proven beneficial in many CVD treatments53. Evidence supports that Ang II increases mitophagy mediator Parkin in VSMCs, which can aid in the removal of damaged mitochondria65. Also in VSMCs, oxidized LDL increases PINK1, ubiquitinated MFN2, and LC3 association to the mitochondria to protect against apoptosis 66. In an experimental model of kidney fibrosis via unilateral ureteral obstruction, MFN2 and Parkin were decreased, while LC3-II was also decreased. Furthermore, both Pink1−/− and Prkn−/− mice display higher collagen deposition in obstructed kidneys compared to WT mice, via enhanced macrophage TGF-β1 expression67. These studies show that deficiencies in the removal of damaged mitochondria can have detrimental effects on cellular death and inflammation. Therefore, suppression of mitochondria damage or enhancing removal pathways via pharmacological tools may yield protection against hypertension.
Mitochondrial DNA Activation of cGAS-STING Signaling
Cytosolic DNA sensing by the cyclic GMP-AMP synthase (cGAS)-STING pathway is implicated in a variety of biological processes associated with intracellular DNA stress. DNA from various sources including the release of mitochondrial DNA (mtDNA) following stress can enter the cytosol and engage the cGAS-STING pathway. Upon activation, STING translocates from the ER to the Golgi, where it recruits kinases TANK- binding kinase 1 (TBK1) and IκB kinase, which phosphorylate interferon regulatory factor 3 (IRF3). Phosphorylated IRF3 dimerizes and translocates to the nucleus to activate transcription of genes encoding type I interferons such as interferon-β (IFNβ)68.
The prime function of this pathway is in host defense via the inflammasome activation. However, recent studies have established a link between CVD stressors and the STING pathway. IRF3 is abundantly found in human samples of coronary plaques69. Phenylephrine-induced aortic contraction is enhanced in Ang II-infused wild-type mice, but not in STING deficient mice, indicating the involvement of STING signaling in aortic contractile dysfunction69. IRF3 reduction is not protective against Ang II infusion-mediated hypertension or cardiac hypertrophy, but it does protect against hypertension-associated cardiac fibrosis70. Saturated fatty acid palmitate induces EC inflammation, an effect that appears to be mediated by mtDNA release and IRF3 activation71. Moreover, mitochondrial damage inducer CCCP and purified mitochondrial DNA increase ICAM1 expression in ECs. Silencing cGAS, STING or IRF3 inhibited palmitic acid-induced mtDNA release and reduced adhesion molecule expression71. According to these findings, the STING/IRF3 signal induced by damaged mitochondria and subsequent mtDNA release may represent drug targets against vascular inflammation and cardiac fibrosis associated with hypertension.
ER-Mitochondria Communication
Mitochondria are linked to the ER at specialized regions of membrane adherence called mitochondria-associated membranes (MAMs), which facilitate Ca2+flux into the ER. Physical linkage between the two organelles occurs via inositol 1,4,5-trisphosphate receptors (IP3R), voltage-dependent anion-selective channel protein (VDAC1) and a chaperone GRP75. Transforming growth factor-β (TGF-β), a key cytokine in hypertension, causes uncoupling of mitochondria from the ER Ca2+ release by suppressing IP3R-mediated Ca2+ efflux72. Furthermore, spatial colocalization of IP3R to TRPC3 channels enhances vasoconstriction in hypertension73. The MAMs and their dynamic regulation seem understudied in hypertension and warrant further investigation. Figure 2 summarizes mitochondrial stress signaling potentially involved in hypertension and associated CVD.
Figure 2. Pharmacological targets of mitochondrial stress signaling in hypertension.

AT1R signaling induces oxidative stress in the mitochondria. In the vasculature, hypertensive stimuli lead to decreased antioxidant mitochondrial components SIRT3 and SOD2, increased ATP production via enhanced FAM3A and ETC complexes. Increased mitochondrial ROS and damage induces adaptative stress responses. Mitochondrial-associated membranes (MAMs) link mitochondria to the ER. Cellular stress causes a surge of Ca2+ via VDAC1 and IP3R which contributes to apoptosis initiation. Mitochondrial damage also leads to the release of mtDNA which can be sensed by cyclic GMP-AMP synthase (cGAS) and its downstream signaling effector stimulator of interferon genes (STING). This leads to NF-κB and IRF3 transcription of interferons and cytokines to contribute to inflammation. Mitophagy via PINK1 depolarization in the inner mitochondrial space leads to Parkin ubiquitin ligase activity on the outer mitochondria membrane which recruits autophagy machinery for phagosome engulfment of damaged mitochondria. Mitochondrial damage also heightens DRP1 GTPase activity to induce fission and tilt the balance away from fusion (mediated via MFN2). Excessive mitophagy and mitochondrial fission has been found in hypertensive models and contributes to vascular pathology. Created with BioRender.com.
Hypertension and Vascular Senescence
As we focus on targeting organelle dysfunction to circumvent hypertensive vascular dysfunction, it is necessary to address other cellular phenotypic changes that organelle homeostasis can mediate. For instance, cell senescence (“to grow old in Latin”) has been characterized in many animal models of CVD74–77, including those with hypertension49, 78–80. In humans, hypertension positively correlates with age, and hypertensive insults may accelerate the cellular aging process including senescence. Therefore, targeting this process may allow an abatement in vascular age acceleration to attenuate aortic stiffening, blood pressure, and end-organ damage81.
Ang II stimulates “stress-induced” premature senescence (SIPS) in VSMCs and mouse aorta, leading to a permanent cell cycle arrest through induction of cyclin dependent kinase inhibitors (CDKi) p16INK4A and p53/p21CIP1 78, 81–83. Senescence in circulating endothelial progenitor cells is increased in SHR, DOCA-salt treated rats and hypertensive humans84. Expression of p16INK4A is increased in heart, kidney and vasculature of DOCA-salt hypertensive rats, which was attenuated by antihypertensive medications85. Arterial telomere uncapping and p21 activity are linked to hypertension independently of mean telomere length in humans86.
Senescent cells evade immune clearance and undergo senescence-associated heterochromatin foci changes, leading to upregulation of anti-apoptotic proteins87. This dictates their ability to reside in tissues for long periods and contribute to organismal dysfunction87, 88. Upregulation of NF-κB signaling occurs in senescent cells and coincides with SASP, with increased secretion of inflammatory cytokines89, 90. SASP has been shown to contribute to frailty and inflamm-aging in other cell systems90. Removing senescent cells via a senolytic drug or genetic clearance reduced atheroma formation and maturation in LDL receptor knockout mice75 and improved vascular relaxation in aged arteries via VSMC sensitization to NO91. At the cellular level, removal of Ang II-induced senescent endothelial cells via a senolytic treatment prevented enhanced leukocyte adhesion92. While findings are still limited regarding the role of senescence and SASP in hypertension81, these findings support the contribution of senescence and SASP to human hypertension.
ER Stress Modulation of Vascular Senescence
ER proteostasis mechanisms decline with chronological age93 and aging is a risk factor for hypertension and vascular stiffening4, 44. ER stress and subsequent UPR induction are associated with all major senescent hallmarks like increased CDKi and β galactosidase expression94. A relationship between ER stress and senescence has been identified in vascular disease. However, the ability to reduce senescent cell accumulation via pharmacological reduction of ER stress remains unclear. In apolipoprotein E−/− mice, VSMCs overexpression of senescence-associated progerin exacerbated atherosclerosis via increased ER stress markers, including GRP78, in the medial layer. Pharmacological inhibition of ER stress via TUDCA ameliorated aortic adventitia-to-media thickness, lipid deposition, and improved survival rate in these mice95. In aged mice, the ER resident chaperone protein involved in Ca2+ handling, calreticulin, is significantly decreased in mesenteric arteries. Calreticulin knockdown impairs Ca2+ signals and endothelial-dependent vasodilation which can be restored with TUDCA treatment in aged mice96. NOX4 gene expression, as well as expression of genes involved in ER stress, was significantly increased in aortas from aged mice. In addition, NOX4-derived ROS controlled endothelial eNOS uncoupling, which was dependent on IRE1α oxidation by sulfonylation97. As mentioned, at the cellular level, 4-PBA treatment prevented Ang II-induced senescence in VSMCs26 as well as ECs92. This represents a novel target since aging induces post-translational modifications of UPR signaling molecules and contributes to vascular dysfunction.
Mitochondrial Dysfunction and Vascular Aging
Reduced mitochondrial biogenesis, reduced mitophagy, increased mtDNA damage, increased mitochondrial ROS with reduced antioxidant defenses are shared hallmarks of both aged and hypertensive vessels98. Oral supplementation with a mitochondrial antioxidant, MitoQ, has been shown to increase NO-mediated endothelial function by 42% over placebo conditions in older adults99. Stress signaling effects of heat shock binding partner-1 (HSB-1) inhibition are mediated via increased histone H4 levels that results in enhanced compaction of both nuclear and mitochondrial genomes. Histone levels decline with age resulting in increased genomic instability, and therefore increased H4 reduce mtDNA damage while simultaneously reducing ETC complex transcription to mitigate excess age-associated mitochondrial oxidative stress100. Senescent cells have hyperfused mitochondria that collect damage. Therefore, enhancing mitophagy clearance mechanisms may reduce vascular senescence. Loss of DRP1 during senescence exacerbates ECs dysfunction by increasing mitochondrial ROS and subsequently inhibiting autophagic flux101. However, in another study inhibition of DRP1 activity via mdiv1 reduces abdominal aortic senescence48.
Responses Against Oxidative Stress
Disruption of the fine balance between free radicals generation and removal is an important cause of cellular stress102–105. Many ROS, including O2.−, H2O2, hydroxyl radical (OH−), peroxynitrite (ONOO−), are highly relevant in oxidative processes that lead to protein inactivation or degradation, reduced NO bioavailability, activation of inflammatory and apoptotic cellular responses105, 106. NADPH oxidase, cyclooxygenases, uncoupled eNOS, xanthine oxidase, and mitochondrial ETC directly contribute to unbalanced redox processes and arterial damage17, 106. Antioxidant defenses are critical in redox-related stresses to prevent ROS-induced oxidative damage and to restore cellular redox homeostasis107.
The Antioxidant Defense Pathway
The nuclear factor (erythroid-derived 2)-like 2 (NRF2) - Kelch-like ECH-associated protein-1 (KEAP1), considered “the” cellular homeostatic guardian, stimulates expression of many antioxidant enzymes. NRF2, encoded by the NFE2L2 gene, is a stress-responsive transcription factor that belongs to cap’n’collar (CNC) subfamily of basic-leucine-zipper (bZIP) transcription factors108. The protective effects of NRF2 against environmental insults was noted when Itoh and colleagues109 observed that electrophile counterattack response is abrogated in NFE2L2-null mice. The electrophile counterattack response is a crucial cellular adaptation108 that protects sulfhydryl groups from oxidation, reduction, or alkylation - events that lead to cell functional loss or death. To activate this response, sulfhydryl reactive electrophiles in specific molecular sensors evoke a coordinated increase in phase II enzymes, such as glutathione S-transferases, epoxide hydratase, AD(P)H: quinone oxidoreductase, and other detoxication enzymes108, 110. NRF2 is the trans-regulatory factor that interacts with the antioxidant responsive element (ARE) and increases the expression of these enzymes108, 111, 112.
The isolation and characterization of KEAP1 clarified how the cell senses and appropriately responds to oxidative attacks113. KEAP1 is a thiol-rich protein that promotes NRF2 degradation in unstressed conditions. Pro-oxidative stimuli directly modify KEAP1 thiols, resulting in KEAP1 inactivation, NRF2 stabilization and induction of cytoprotective genes via ARE114, 115. KEAP1 is therefore the direct biosensor for electrophiles and ROS unbalance, while NRF2 is the molecular effector that maintains cellular redox homeostasis.
NRF2 Pathway, Hypertension and CVD
Pharmacological activation of NRF2 by tert-butylhydroquinone prevents Ang II-induced hypertension, microvascular remodeling, and endothelial dysfunction in wild-type, but not in Nrf2-deficient mice116. The importance of NRF2 activation in hypertension-associated vascular dysfunction is further supported by studies showing that SHRSP have reduced NRF2 activity and NRF2-targeted gene expression, events linked to the vascular dysfunction exhibited by these animals. NRF2 activators (bardoxolone/sulforaphane) improve SHRSP’s vascular function and reduce Ang II-induced ROS generation in VSMCs117. NRF2 activation by sulforaphane maintains a healthy endothelial phenotype under proinflammatory conditions118, whereas disruption of Nrf2 signaling impairs angiogenic responses in ECs119. In two kidney-one clip (2K-1C) rats, oral nitrite administration increases NRF2 activation and expression of NRF2-regulated genes (SOD1, catalase, glutathione peroxidase, thioredoxin-1 and −2) and attenuates vascular dysfunction120, suggesting that NRF2 activation contributes to the antioxidant effects of oral nitrite.
Selective Nrf2 gene deletion in the rostral ventrolateral medulla (RVLM) increases sympathetic outflow and impairs baroreflex function, inducing hypertension, potentially by disrupting antioxidant enzyme expression121. Of clinical importance, single nucleotide polymorphism in NFE2L2 (rs6721961 CA+AA) and KEAP1 (rs110857735 the AA or CA) is linked to altered blood pressure and adverse cardiovascular outcomes in Japanese122 and Italian123 patients, respectively.
Interestingly, Ang II-infused Nrf2−/− mice exhibit exaggerated cardiac hypertrophy with no difference in the development of hypertension compared to wild-type mice124. Despite evidence supporting an anti-hypertensive role for NRF2 activation, enhancement of NRF2 activity via genetic inhibition of KEAP1 appears rather harmful, since it leads to higher blood pressure upon Ang II infusion125.
NRF2 also prevents vascular damage in diabetes, a condition that increases the risk of hypertension. Diabetic db/db mice exhibit decreased renal NRF2 activity, downregulation of antioxidant enzymes and nephropathy, events that are mediated the adipokine chemerin and its receptor ChemR23126. Impaired NRF2 signaling can be associated with disrupted Akt-dependent signaling127. Although KEAP1 is key to regulate NRF2 activity in response to electrophiles, NRF2 is also activated via PI3K-AKT signaling128. Akt induces NRF2 activity129 primarily by inhibiting GSK-3, glycogen synthase kinase-3β, which phosphorylates NRF2 and marks it to ubiquitination and degradation130. In streptozotocin-induced diabetic mice, Nrf2 activation by L-sulforaphane or bardoxolone rescues internal pudendal artery function131, suggesting that this antioxidant system may be important in numerous vascular dysfunction conditions, as shown in Table 1. In this context, understanding NFR2 signaling in physiological and pathological conditions not only clarifies its function in redox balance, but also contributes to unravel potential therapeutic strategies targeting vascular dysfunction.
NRF2 in Proteostasis and Senescence
NRF2 not only protects from oxidative stress, but also modulates general adaptive cellular processes, such as inflammation132, metabolic programming133 and proteostasis134. In vascular stress, NRF2 is activated via PERK phosphorylation, as a protective mechanism to limit CHOP-induced apoptosis in ECs135. NRF2 pharmacological inhibition with ML385, in a myocardial infarction mouse model, suppresses cardioprotective effects of tumor susceptibility gene 101 (Tsg101), which promotes p62 aggregation, KEAP1 degradation and NRF2 translocation to the nucleus136. This highlights a link between proteostasis and NRF2 antioxidant responses. In senescent vascular cells, the lack of an effective NRF2 response to oxidative stress results in overwhelming ROS levels137, 138. Nrf2 null mice have higher levels of IL-1β and TNF-α, well-known components of the SASP. Additionally, Nrf2 deletion increases the expression of molecular senescence markers (p16INK4a, p21) in cerebral arteries of 24-month-old mice139. Endothelial Nrf2 mRNA is reduced by miRNAs derived from senescent cells, such as miR-126, miR-21, and miR-100140, further reinforcing that NRF2 is also a potential therapeutic target to treat aging-related diseases141. Figure 3 illustrates how NRF2 modulates vascular (dys)function.
Figure 3. NRF2 and oxidative stress.

Impaired NRF2 activation reduces the expression of antioxidant proteins, leading to exacerbation of cellular oxidative damage. In endothelial cells, ROS overproduction reduces NO bioavailability, impairs vasodilation, causes inflammatory endothelial activation, ER stress and mitochondrial dysfunction. In VSMCs, oxidative damage also activates proliferative pathways and increases vasoconstrictor responses. NADPH Oxidase (NOX), Toll-like receptors (TLR), angiotensin II (Ang II), nitric oxide (NO), nuclear factor (erythroid-derived 2)-like 2 (NRF2), Kelch-like ECH-associated protein 1 (KEAP1), antioxidant response element (ARE), Small Maf proteins (sMAF), damage-associated molecular patterns (DAMPs), pathogen-associated molecular patterns (PAMPs). Created with BioRender.com.
NRF2-inducers/activators, such as bardoxolone (government identifier: NCT00811889; NCT01351675)142, and sulforaphane (government identifier: NCT01008826; NCT02880462; NCT02801448; NCT03220542)143 are currently being tested in clinical trials. Other natural NRF2 activators, such as polyphenols, phytosterols, and terpenoids (e.g., Baicalein), are also being tested in pre-clinical models of kidney disease144. So far, data from these studies suggest that modulation of the NRF2-KEAP1 pathway is a potential therapeutic strategy to combat oxidative stress-induced cellular damage. This raises the possibility of successful results also in CVD. However, contrasting results have been reported. While bardoxolone improves estimated glomerular filtration rate in chronic kidney disease patients142 and sulforaphane improves glucose metabolism in patients with dysregulated type 2 diabetes143, bardoxolone failed to show positive effects in patients with type 2 diabetes and end-stage chronic kidney disease145. The negative or unsuccessful result in this specific study may be linked to the high-risk group of patients enrolled in the trial.
Although countless preclinical studies show detrimental roles of ROS and oxidative stress in the cardiovascular system as well as beneficial cardiovascular effects of antioxidants, many clinical trials with antioxidant compounds [e.g. with β-carotene, vitamin E, vitamin C, α-lipoic acid, polyphenols (epigallocatechin gallate, curcumin, resveratrol) and flavonoids (quercetin, rutin)] have not confirmed beneficial or protective effects144. In many cases, adverse effects such as cardiac ischemia, intracerebral hemorrhage, increased risk of heart failure, decreased vasodilation, and oxidative damage, were reported. Adverse effects are usually observed with high doses of antioxidants and many studies show that antioxidants exhibit dose-dependent effects102. Potential possibilities to explain the negative results in the clinical trials include: i) antioxidant drugs may abrogate physiological and relevant oxidant processes; ii) antioxidants reduce metal ions, such as Fe3+ and Cu2+, favoring the formation of other free radicals (•OH, e.g.); iii) antioxidants may not distinguish between radicals involved in physiological processes and those that cause oxidative damage; iv) removal of oxidants may disrupt essential endogenous antioxidant responses (e.g. NRF2 signaling) and thus decrease endogenous antioxidant protection. These hypotheses led to terms such as reductive stress, antioxidative stress and antioxidant paradox. Additional possibilities include v) antioxidants may modify macromolecules, which by unrelated mechanisms negatively impact beneficial effects; vi) antioxidant effects may be hampered by inadequate duration of treatment, poor absorption and/or weak scavenging activity. The beneficial vascular effects of physiological ROS and how excessive removal of ROS might perturb vascular homeostasis was recently reviewed102.
Many authors argue that antioxidants should be target-specific – i.e. directed to selective inhibition of ROS-generating enzymes (e.g. NADPH oxidase isoforms) or to activation of specific antioxidant signaling (such as NRF2 and specific antioxidant enzymes)146. Others consider that antioxidants should be given in low doses in order to avoid suppression of physiological ROS; or that administration of low levels of pro-oxidants may enhance the antioxidant capacity of cells102, 146. Finally, inclusion of specific groups of patients in the clinical trials should be considered, since some of the negative results were obtained in very ill patients, including patients with end-stage chronic kidney disease plus type 2 diabetes145. Studies on all these topics are still incipient but may reveal new strategies against the detrimental effects of oxidative stress in hypertension and CVD.
Signaling Via Pattern Recognition Receptors
Pattern recognition receptors (PRRs), i.e. proteins found in the cell membrane, in subcellular compartments, in the cytosol, and extracellularly, recognize molecules expressed by pathogens (the so-called pathogen-associated molecular patterns – PAMPs) or molecules released by damaged cells (the damage-associated molecular patterns – DAMPs). Four major sub-families of PRRs are known: Toll-like receptors (TLRs), nucleotide-binding oligomerization domain (NOD)- Leucine Rich Repeats (LRR)-containing receptors (NLR), retinoic acid-inducible gene 1 (RIG-1)-like receptors (RLR), and C-type lectin receptors (CLR). PRRs not only activate pro-inflammatory responses, but also activate and regulate cell death signaling pathways147–149. TLR, for instance, can trigger apoptosis, while inflammasomes can activate key proteins in necroptotic cells, as well as caspase-11, important for pyroptosis150. Of note, in a clinical trial for patients with refractory solid tumors, the use of a vaccine containing NOD2 and TLR9 stimulants worsened arterial hypertension from grade 2 to grade 3 in 20% of the patients, indicating their role on rising blood pressure151. Thus, due to solid evidence linking TLRs and NLRP3 inflammasome to arterial hypertension and CVD, in this last section these critical PRRs will be discussed.
Toll-Like Receptors
The discovery of TLRs as PRRs changed the idea that the innate immune system is a simple mechanism to initiate the adaptative immune response, shedding light for a broader understanding of the innate immune system in health and disease152. TLRs are expressed in immune and nonimmune cells. In the cardiovascular system, TLRs expressed in ECs and VSMCs sense the danger in the advent of a pathogen invasion to trigger the inflammatory response and control the local blood flow 153. As shown in Figure 4, TLRs are also activated by DAMPs, which are fragments of damaged or dying cells that could cause a chronic low-grade inflammation by prolonged or excessive activation of the TLRs, causing endothelial dysfunction and CVD154.
Figure 4. Ligands for TLRs and NLRP3 inflammasome.

TLRs and NLRP3 inflammasome are involved in a growing number of infectious, autoimmune, and metabolic diseases. Nearly every class of microbe and many cell-derived as well as synthetic compounds activate or modulate TLRs and NLRP3 inflammasome. The Hemozoin, disposal product formed from the digestion of blood by some blood-feeding parasites such as Plasmodium. Nigericin, microbial toxin derived from Streptomyces hygroscopicus. Flagellin, structural component of the bacterial flagellum. PFT, pore-forming toxins. ATP, adenosine 5’-triphosphate. Drusen, aging- and macular degeneration-associated accumulations of extracellular material that build up between Bruch’s membrane and the retinal pigment epithelium of the eye. Nano-SiO2, silica dioxide nanoparticles.
At least 10 functional TLRs have been identified in human cells. TLR1, TLR2, TLR5, TLR6 and TLR10 are expressed in the plasma membrane of the cells, while TLR3, TLR7, TLR8, TLR9 are expressed in the endosomes and ER and, lastly, TLR4 can be found in both plasma membrane and endosomal vesicles155. In rodents, TLR11, TLR12 and TLR13 are also described156, 157. Nonetheless, TLR activation is mediated by specific ligands and the downstream pathway of TLRs is associated with different adaptor proteins and ultimately leading to the production of proinflammatory cytokines and interferons. The role of TLRs in hypertension is not fully elucidated. However, there is evidence of the contribution of TLRs for endothelial and vascular dysfunction leading to hypertension. Figure 5 illustrates the downstream pathway in TLR activation.
Figure 5. Activation of TLRs pathway and their downstream targets.

In hypertension, TLRs expressed in the cell membrane or in the endosomes are activated by damage-associated molecular patterns (DAMPs) derived from the cellular damage and it triggers the downstream signaling pathway through the activation of adaptor molecules myeloid differentiation protein (MyD88) and TIR domain-containing adaptor protein inducing interferon-β (TRIF) to induce the activation of NF-κB, which will, ultimately, lead to the formation of proinflammatory cytokines and interferons.
Among the TLRs, activation of TLR2 (which is known for heterodimerizing with TLR1 or TLR6 to recognize triacyl or diacyl lipopeptides, respectively) was shown to play a role in the regulation of the vascular tone. Specifically, vessels exposed to pathogens exhibited a diminished contractile force, which was not observed in TLR2−/− mice. Activation of TLR2 is associated with inducible nitric oxide synthase (iNOS)-derived NO formation, in a mechanism dependent on heterodimerization of TLR2 with TLR6 and subsequent activation of the adaptor protein MyD88 (myeloid differentiation protein), MAL (MyD-adapter-like protein) and TNF-α production158. In addition, treatment with Ang II led to the development of fibrosis in the aorta as well as endothelial to mesenchymal transition (EndMT) via NF-κB signaling in human umbilical vein ECs, through activation of TLR2, and these effects were prevented by knocking out or silencing TLR2159. Although activation of TLR2 by pathogens led to an anticontractile effect on the vessels, damage-associated activation of TLR2 seems to contribute to endothelial dysfunction and the development of hypertension. Increased systolic blood pressure (SBP) was observed in mice injected with abnormal HDL containing symmetric dimethylarginase (HDL+SDMA) obtained from patients with chronic kidney disease (CKD). However, injection of HDL+SDMA in TLR2−/− mice (but not in TLR4−/−) failed to increase SBP, supporting the idea that the increase in SBP was mediated by activation of TLR2160. In ECs, HDL+SDMA decreased NO bioavailability and increased ROS, however, by a mechanism independent of TLR1 or TLR6 dimerization160. In addition, in vascular smooth muscle, dysfunctional HDL from mice with CKD or humans with end-stage renal disease induced the mRNA expression of MCP-1 (monocyte chemoattractant protein 1) through activation of TLR2 and TLR4161.
Indeed, evidence has shown that TLR4 also plays a role in endothelial and vascular dysfunction in several pathological conditions, including diabetes162 and sepsis163. Classically, TLR4 is the specific sensor for lipopolysaccharide and its activation leads to the recruitment of the adaptor protein MyD88/MAL or TRIF (TIR domain-containing adaptor protein inducing interferon-β; TIR: Toll/IL-receptor domain), to produce several proinflammatory cytokines or interferons, respectively164. In addition, high mobility group box 1 (HMGB1), an alarmin and well recognized as a member in SASP, has been highly associated with the activation of TLR4 and receptor for advanced glycation end-products (RAGEs). HMGB1 and TLR4 activation is correlated with pulmonary hypertension165 and preeclampsia166. Activation of TLR4 also plays a role in the development of vasculogenic erectile dysfunction, which is often considered an early marker for CVD167. Although TLR4 is less investigated in arterial hypertension, administration of VIPER (a specific TLR4 blocker) in the paraventricular nucleus (PVN) of SHR for 14 days, reduced mean arterial pressure (MAP) and expression of TLR4 in the PVN and plasma. Additionally, VIPER reduced PVN levels of HMGB1, and decreased expression of TNF-α, IL-1β, iNOS while increasing expression of IL-10168. Similarly, in two-kidney, one-clip (2K1C) hypertensive rats, physical exercise attenuated the increase in SBP and reduced expression of TLR4, and its downstream effectors MyD88, NF-κB, TNF-α and IL-1β in the PVN with the same magnitude as the TLR4 inhibitor TAK242169. In addition, SHR treated with losartan or an herbal product displayed reduced expression of TLR4, MyD88, NF-κB, TNF-α and IL-6, which was accompanied by an improvement in cardiovascular parameters (SBP, DBP, MAP, HR and flow), prevention of cardiac hypertrophy and fibrosis and preserved endothelium-dependent vascular relaxation170. Similarly, SHR exhibited increased expression of TLR4 in mesenteric arteries and treatment with anti-TLR4 decreased blood pressure and it reduced the expression of TLR4, cyclooxygenase-2 (COX-2) and thromboxane A2. The contractile response to norepinephrine in the mesenteric artery, as well as the circulating levels of IL-6171 were also normalized after treatment with anti-TLR4. In humans, HMGB-1 is increased in the plasma of patients with peripheral artery disease and it is positively correlated with the severity of PAD172. Furthermore, endothelial dysfunction is associated with increased levels of HMGB1 in women with polycystic ovary syndrome173. Despite the correlation of TLR4 with vascular complications in different diseases and contrary to what has been observed in SHR, Ang II infusion in TLR4−/− mice did not prevent the increase in the SBP, although the cardiac hypertrophy was attenuated174. Thus, the role of TLR4 in hypertension is still controversial. On the other hand, infusion of Ang II in TLR3−/− mice prevented the increase in the SBP as well as the cardiac hypertrophy174. TLR3 is localized in the endosomes and senses viral nucleic acids or mitochondrial double-stranded RNA released from damaged cells. Its downstream signaling pathway occurs exclusively through TRIF175. Interestingly, many viral infections are related to increased blood pressure or hypertension, therefore, activation of TLR3 could be the missing link between these conditions5. Corroborating this idea, TLR3 activation is associated with endothelial dysfunction and increased ROS176 and it is also correlated with increased blood pressure in animal models of preeclampsia177. Activation of TLR3 in human microvascular ECs causes augmented expression of pro-inflammatory cytokines and damages to the endothelial barrier integrity, which are potentiated when eNOS is silenced or inhibited with L-NAME178. Although the inflammation caused by activation of TLR3 does not play a role in the development of arterial hypertension, it is possible that TLR3 activation worsens the endothelial function in hypertension, by decreased expression/activity of eNOS.
Similar to TLR3, TLR9 is expressed in endosomes of immune and nonimmune cells, and it is activated by unmethylated DNA. Upon activation of TLR9, the adaptor protein MyD88 forms a complex with IL-1-receptor-activated kinase (IRAK4) and TNF receptor-associated factor (TRAF6), to induce the activation of NF-κB and generate proinflammatory cytokines155. In SHR, impaired autophagic machinery led to increased circulating levels of mtDNA, activating TLR9 and leading to vascular dysfunction179 whereas inhibition of TLR9 in young SHR (but not in adult SHR) led to a milder increase in blood pressure and recruitment of immune cells to the vessels when compared to young SHR with intact TLR9180. In addition, adult male SHR exhibit higher SBP compared to female SHR, and likewise, circulating levels of mtDNA and pro-inflammatory cytokines, as well as contractile responses to phenylephrine are higher in male SHR than in female SHR181, reinforcing the idea that the increase in blood pressure is mediated by TLR9 activation. Furthermore, endothelial/vascular dysfunction associated with TLR9 activation is also observed in other pathological conditions such as prediabetes182, preeclampsia183 and atherosclerosis184.
Other TLRs have been reported to contribute to endothelial and/or vascular dysfunction in some cardiovascular complications/diseases, however, not in arterial hypertension. It is known that TLR7 and TLR8 activate downstream signaling pathways similarly to TLR9, through NF-κB release and formation of the proinflammatory cytokines TNF-α, IL-6, IL-1β, IL-12 and type 1 interferon185. The implication of TLR7 and TLR8 for CVD are not well established. However, a recent study showed that, in mice, the topical administration of imiquimod to induce lupus through TLR7 activation also induced hypertension, endothelial dysfunction, decreased eNOS expression and caused vascular remodeling, associated with increased levels of proinflammatory cytokines186. In contrast, another study showed that the activation of TLR7 (as well as TLR3 and TLR8) leads to an increase in blood pressure and endothelial dysfunction only in pregnant mice and it was also found that the levels of these TLRs were increased in the placenta of women with preeclampsia187. Together, this can be indicative that the activation of TLR7 (and 8) does not trigger the development of vascular complications, unless upon the existence of another inflammatory condition. As for TLR8, its activation seems to be involved in the development of atherosclerotic plaques in rabbits fed high lipid diet188. In addition to the inflammatory state observed by overactivation of the TLRs in these pathological conditions, further mechanisms have been demonstrated that could link TLRs and hypertension.
RhoA/Rho-kinase (ROCK) signaling is critical in arterial contraction and hypertension. ROCK negatively regulates the myosin light chain phosphatase subunit, myosin phosphatase target subunit 1 (MYPT1), a physiological ROCK substrate. Thus, ROCK activation causes Ca2+ sensitization in VSMCs, favoring the contractile state, and its overexpression/activity is largely associated with hypertension189. Interestingly, studies have reported a communication between some TLRs and activation of RhoA/ROCK signaling in the vasculature and other organs, such as in the corpus cavernosum, where activation of TLR1/2 increases contraction and impairs relaxation to Y-27632, suggesting that TLR1/2 activation leads to increased ROCK signaling190. Similarly, a noncanonical pathway related to the activation of TLR9 has been described and it could be involved in the underlying mechanism of TLR9-induced hypertension. In VSMCs, treatment with the TLR9 agonist ODN2395, induced vascular autophagy by activation of the AMPK pathway via TGF-β–activated kinase 1 (TAK1), as well as phosphorylation of MYPT1 and cofilin, leading to actin polymerization and impaired arterial relaxation191. Interestingly, restoration of arterial autophagy in SHR improved remodeling and endothelium-dependent relaxation of the mesenteric resistance artery and reduced the expression of ROCK and phosphorylation of MYPT-1, preventing the early vascular aging in hypertension44. Overall, activation of different TLRs can trigger mechanisms associated with the development of hypertension (Table 1). Nevertheless, TLRs are not the only PRRs that may be involved in this process.
NLRP3 Inflammasome
Certain members of the PRRs are assembled in complex structures or platforms called inflammasomes. Canonical inflammasomes are composed of a sensor protein, usually a NLR or a pyrin and HIN domain-containing protein (PYHIN), an adaptor molecule, the apoptosis-associated speck-like protein containing a caspase activating and recruitment domain (ASC), and pro-caspase-1. Activation of the cytosolic sensors in response to cell stress [represented by PAMPs, DAMPs or cytosolic instabilities (ionic imbalance, oxidative stress)] leads to the recruitment and activation of caspase-1 either directly or through the ASC adaptor molecule. Among the five major types of inflammasomes [NLR pyrin domain-containing 3 (NLRP3), NLRP1, neuronal apoptosis inhibitory protein (NAIP)/NLR CARD-containing 4 (NLRC4), absent in melanoma 2 (AIM2), and PYRIN], NLRP3 has been linked to uncontrolled inflammation in many CVD, including arterial hypertension192, 193.
NLRP3 inflammasome activation involves two coordinated steps, as shown in Figure 6. In the first step (priming step), DAMPs or PAMPs-induced activation of TLRs lead to NF-kB signaling, synthesis of pro-IL-1β, pro-IL-18 and increased expression of NLRP3 components. The second step involves NLRP3 inflammasome assembly, which triggers the self-cleavage and activation of caspase-1, and conversion of pro-IL-1β and pro-IL-18 to their mature forms194. An intriguing and not yet resolved point refers to the mechanisms linking the priming and activation steps. Distinct stimuli can trigger this process, including pathogens, K+ efflux, increased intracellular Ca2+, lysosomal leakage, mitochondrial damage and ROS (derived from ER stress, damaged mitochondria and NADPH oxidase)194–196. ER stress activates NF-κB, thioredoxin-interacting protein (TXNIP) and sterol regulatory-element binding proteins (SREBPs) signaling, Ca2+ release, and ROS production. However, a common intracellular intermediate that leads to NLRP3 assembly and activation has not been identified. Several kinases and phosphatases modify NLRP3 in response to either PAMPs or DAMPs and provide fine adjustments of NLRP3 activation150, 197.
Figure 6. Two step model for NLRP3 inflammasome activation.

In hypertension and aging, the priming signal (first signal) is provided by proinflammatory cytokines, such as IL-1β and TNF, or molecules released from damaged cells (the so called damage-associated molecular patterns – DAMPs) that interact with specify membrane receptors (IL-1R, TNFR, TLR) leading to activation of transcription factor nuclear factor-kappa B signaling. NF-κB upregulates gene expression of NLRP3 components (NLRP3, ASC adaptor molecule), pro-caspase 1, pro-IL-1β, pro-IL-18 and other pro-inflammatory mediators. The complete NLRP3 assembly is induced by a second signal, that can be represented by a plethora of factors - K+ efflux, increased intracellular Ca2+, lysosomal leakage, mitochondrial damage and ROS mainly derived from ER stress, damaged mitochondria [(mt)ROS and mtDNA], and NADPH oxidase (NOX). ER stress also activates NF-κB, TXNIP, and SREBP signaling, further contributing to NLRP3 activation. Activated NLRP3 recruits and activates caspase-1 that cleavages pro-IL-1β, pro-IL-18 and FL-GSDMD (Gasdermin D) to their respective active forms. GSDMD induces pyroptotic cell death and IL-1β and IL-18 contribute to hypertension- and aging-associated vascular damage. Created with BioRender.com.
IL-1β and IL-18 receptors (IL-1R and IL-18Rα, respectively), are found on several leukocyte subsets, ECs and VSMCs198, 199. Activation of IL-1R and IL-18Rα leads to recruitment of distinct accessory and adapter molecules that facilitate the activation of JNK and p38 MAPK signaling and transcription factors such as activator protein-1 (AP-1) and NF-κB200. These receptors and factors stimulate pro-inflammatory gene expression201, 202 and are relevant to hypertension-associated vascular dysfunction203, 204 and remodeling201, 205–207. In a feedback loop, IL-1β /IL-18 signaling stimulates ROS production by NADPH oxidase206 and COX-dependent mechanisms204 and, consequently, induces NLRP3 overactivation.
Overactivation of the NLRP3 inflammasome has been linked to the development of arterial hypertension and CVD. Omi et al., by examining 1911 patients (987 with hypertension, 924 controls), reported that homozygotes of 12 repeat allele, whose NLRP3 inflammasomes produce more chemokines after stimulation, exhibit greater risk for the development of hypertension compared to heterozygotes or homozygotes of non-12 repeat allele (who exhibit low NLRP3 activity). In addition, mean systolic blood pressure of male homozygotes of 12 repeat allele (high NLRP3 activity) was 6.4 mmHg higher than that of homozygotes of low NLRP3 activity alleles208. Furthermore, hypertensive patients, with hyperaldosteronism and with normal aldosterone levels, exhibit increased NLRP3 inflammasome activation209.
IL-1β and IL-18, end-products of NLRP3 activation, are increased in hypertensive patients205, 210. A recent clinical trial (Canakinumab Anti-inflammatory Thrombosis Outcome Study – CANTOS) showed that canakinumab, a human monoclonal antibody targeting IL-1β, reduces rates of recurrent cardiovascular events211, 212 and hospitalization for heart failure213 in patients with a history of myocardial infarction and persistent pro-inflammatory response. In this context, treatment of DOCA/salt-hypertensive mice with anakinra, an IL-1R antagonist, reduces blood pressure and renal fibrosis, although it does not change kidney leukocyte infiltration214. Although it is clear that IL-1β, the main effector of NLRP3, is important in the development of CVD, including arterial hypertension, the complex mechanisms involved in IL-1β effects on hypertension need further investigation.
NLRP3 blockade also prevents hypertension-associated cardiovascular dysfunction, oxidative stress and structural abnormalities. NLRP3 gene silencing attenuates hypertension, vascular remodeling, and VSMCs phenotype switching in SHR193. Nlrp3 deletion in mice prevents Ang II-induced gestational hypertension and IL-6 up-regulation in the placenta and kidneys215. Additionally, the NLRP3 inflammasome, through activation of IL-1R, mediates aldosterone-induced vascular damage209 and MCC950, a selective NLRP3 inhibitor216, reduces blood pressure and attenuates renal damage and dysfunction in salt-sensitive hypertension192. Of note, MCC950 failed to improve vascular function in aged mice with Ang II-induced hypertension217. Salt-sensitive hypertensive rats exhibit increased IL-1β and NLRP3 in the hypothalamic paraventricular nucleus (PVN). Bilateral injection of gevokizumab (IL-1β inhibitor) into the PVN decreased mean arterial pressure and heart rate in these animals, suggesting that NLRP3 / IL-1β signaling contributes to sympathoexcitation during development of salt-dependent hypertension218.
NLRP3 inflammasome also contributes to vascular damage in diseases closely linked to hypertension, such as atherosclerosis and diabetes. In these conditions, activation of the NLRP3 inflammasome mediates pyroptosis, increases mitochondrial ROS in ECs219 and induces endothelial dysfunction220. NLRP3 inhibitors, such as tranilast and oridin, display anti-atherosclerotic properties by decreasing vascular and endothelial inflammation, restraining activation of MAPK and NF-κB signaling and decreasing cell migration and angiogenesis221–223. Blockade of NLRP3 activation also restores EC function and reduces oxidative stress and inflammatory processes in type 2224 and type 1225 diabetes. In addition, NLRP3 inflammasome is linked to aging-associated dysregulation of the immune system226, endothelial senescence227, and cardiovascular remodeling and dysfunction226, 228. NLRP3 components gene expression is increased in peripheral blood mononuclear cells of older patients with vascular diseases in comparison to younger patients229.
Overall, these studies provide evidence that NLRP3 inflammasome activation contributes to proinflammatory signaling, leading to vascular dysfunction and remodeling and a premature senescence phenotype in hypertension and other CVD. The impact of genetic and pharmacological control of NLRP3 activity on vascular (dys)function is summarized in Table 1. Since NLRP3 assembly and signaling involve many molecules, NLRP3 inflammasome inhibition can be achieved by interfering with upstream molecules (P2X7 receptor, K+ channels, ROS), inflammasome components (NLRP3 domains - e.g. NLRP3 ATP-binding domain -, ASC), and downstream molecules (caspase-1, IL-1β and IL-18 and their receptors), as reviewed by other authors216, 223.
Many questions remain unanswered, demanding further studies: i) which molecules function as cellular stress-signals to trigger NLRP3 activation in hypertension?, ii) what specific signaling pathways are activated when NLRP3 senses different cellular stress signals?, iii) how modulators of cellular redox status (pro- and anti-oxidant enzymes, ER, mitochondria) influence NLRP3 activity?, iv) do current anti-hypertensive medications impact NLRP3 activation and signaling?, v) does simultaneous blockade of NLRP3 inflammasome and IL-1β/IL-18 signaling better prevent hypertension and associated complications?, vi) how does activation of other stress signaling pathways impact NLRP3 activation (and vice-versa)?, vii) do other inflammasomes, such as NLRP1, NLRC4, AIM2, and noncanonical inflammasomes also contribute to hypertension-associated vascular dysfunction?. Another aspect to be explored is that not only immune cells express NLRP3. The inflammasome is also found in myocardial, VSMCs, EC and fibroblasts. NLRP3 inflammasome activation in these cells also contributes to proinflammatory signaling, leading to vascular dysfunction and remodeling and premature senescence phenotype233, 234.
Altogether, the NLRP3 inflammasome, a sensor for danger signals and one of the earliest components activated in inflammatory responses, may represent a critical checkpoint that interconnects pathophysiological events in hypertension. Effective regulation of NLRP3 may help prevent the development and aggravation of hallmark processes in arterial hypertension and CVD, especially in conditions where current available drugs are insufficient to prevent the development or complications of these diseases.
Summary and Conclusion
Vascular changes that contribute to elevated arterial blood pressure in hypertension include the following: endothelial dysfunction, increased smooth muscle contraction, remodeling of the vascular wall, inflammation and increased arterial stiffness. This review provided a synopsis of several stress signaling mechanisms that contribute to these vascular changes and appraised literature indicating that several of these abnormalities occur during the aging process. Common cellular mechanisms involved in vascular dysfunction in hypertension and aging include ER stress and accumulation of protein aggregates, oxidative injury, damage to mitochondria, DNA damage, cell senescence, fibrosis, TLR activation and inflammasome formation. Adaptive mechanisms against these stresses include the UPR, autophagy, ARE signaling, mitophagy and mitochondrial fusion, induction of STING and senolysis. There are several major gaps in our knowledge in terms of developing pharmaceuticals to target these adaptive mechanisms. As these stress responses are all critical in cell homeostasis and defense, unappreciated side effects are expected for any potential target. From a pharmacological standpoint, future studies should consider the following: establishment of controlled targeting for maladaptive stress signaling, better use of potential translational models for screening of the targeting compounds, and investigation on the impact of lifestyle choices and environmental factors on these maladaptive stress signals and their contribution to hypertension.
Acknowledgments
Sources of funding
Financial support for this work was provided by the NIDDK Diabetic Complications Consortium (DK076169 and DK115255 to RCW), the National Institutes of Health (HL-134604 to RCW, HL-133248, DK-111042, NS-109382 to SE and T32 HL-91804 to SC) and The Sao Paulo Research Foundation [(FAPESP) 2016/16207–1 to JFS and 2013/08216–2 to RCT].
Nonstandard Abbreviations and Acronyms:
- 4-PBA
4-phenylbutyric acid
- AAA
abdominal aortic aneurysm
- AIM2
absent in melanoma 2
- AMPK
AMP-activated protein kinase
- Ang II
angiotensin II
- ARE
antioxidant response
- ASC
caspase activating and recruitment domain
- AT1R
Ang II type 1 receptor
- AT2R
Ang II type 2 receptor
- ATAAD
ascending thoracic aortic aneurysm and dissection
- ATF6
activating transcription factor 6
- ATM
ataxia telangiectasia mutated
- ATP
adenosine triphosphate
- ATP
adenosine 5’-triphosphate Nano-SiO2, silica dioxide nanoparticles
- BHB
β-Hydroxybutyrate
- bZIP
basic-leucine-zipper
- Ca2+
calcium
- CANTOS
Canakinumab Anti-inflammatory Thrombosis Outcome Study
- CC
corpora cavernosa
- CDKi
cyclin dependent kinase inhibitors
- CHOP
C/EBP homologous protein
- CKD
chronic kidney disease
- CLR
C-type lectin receptors
- CNC
cap’n’collar
- COX-2
cyclooxygenase-2
- CVD
cardiovascular diseases
- DAMPs
damage-associated molecular patterns
- DRP1
dynamin-related protein
- ECs
endothelial cells
- EndMT
endothelial to mesenchymal transition
- eNOS
endothelial nitric oxide synthase
- ER
endoplasmic reticulum
- ERAD
ER-associated degradation
- ESRD
end-stage renal disease
- ETC
electron transport chain
- FAM3A
family with sequence similarity 3 member A
- FL-GSDMD
(Gasdermin D inactive)
- GRP78
glucose-regulated protein 78
- GSK-3
glycogen synthase kinase-3β
- GTPases
guanosine triphosphatases
- H2O2
hydrogen peroxide
- HMGB1
high mobility group box 1
- HO-1
heme oxygenase-1
- HSB-1
heat shock binding partner-1
- HSP90
heat shock protein 90
- I/R
ischemia/reperfusion
- IFNβ
interferon-β
- IL-1β
interleukin-1β
- IL-6
interleukin-6
- iNOS
inducible nitric oxide synthase
- IP3R
inositol 1,4,5-trisphosphate receptors
- IRAK4
IL-1-receptor-activated kinase
- IRE1α
inositol-requiring transmembrane kinase 1 alpha
- IRF3
interferon regulatory factor 3
- JNK
c-Jun N terminus kinase
- K+
potassium
- KEAP1
Kelch-like ECH-associated protein 1
- LRR
Leucine Rich Repeats
- MAL
MyD-adapter-like protein
- MAMs
mitochondria-associated membranes
- MAP
mean arterial pressure
- MCP-1
monocyte chemoattractant protein 1
- MFN1 and MFN2
mitofusins 1 and 2
- miR
microRNA
- MLKL
mixed lineage kinase domain-like
- mtDNA
mitochondrial DNA
- mtROS
mitochondrial-derived ROS
- MyD88
myeloid differentiation protein
- MYPT1
myosin phosphatase target subunit 1
- NOX
NADPH Oxidase
- NAIP
neuronal apoptosis inhibitory protein
- NF-κB
nuclear factor-κB
- NLR
containing receptors
- NLRC4
NLR CARD-containing 4
- NLRP3
NLR pyrin domain-containing 3
- NO
nitric oxide
- NOD
nucleotide-binding oligomerization domain
- NOX
HADPH oxidase
- Nrf2
nuclear factor (erythroid-derived 2)-like 2
- O2.-
superoxide anion
- OH-
hydroxyl radical
- ONOO−
peroxynitrite
- OPA1
optic atrophy 1
- ox-LDL
oxidized LDL
- p38 MAPK
p38 mitogen-activated protein kinases
- PAMPs
pathogen-associated molecular patterns
- PRRs
pattern recognition receptors
- PERK
protein kinase-like ER kinase
- PFT
pore-forming toxins
- pro
IL-18
- pro
IL-1β
- PVN
paraventricular nucleus
- PYHIN
pyrin and HIN domain-containing protein
- RAGE
receptor for advanced glycation end-products
- RhoA
Ras homolog family member A
- RLR
inducible gene 1 (RIG-1)-like receptors
- ROCK
RhoA/Rho-kinase
- ROS
reactive oxygen species
- RVLM,
rostral ventrolateral medulla
- SAHF
senescence-associated heterochromatin foci
- SASP
senescence-associated secretory phenotype
- SBP
systolic blood pressure
- SHR
spontaneously hypertensive rats
- SHRSP
stroke-prone spontaneously hypertensive rats
- SIRT3
mitochondrial sirtuin 3
- sMAF
small Maf proteins
- SODs
superoxide dismutases
- STING
signaling effector stimulator of interferon genes
- TAK1
transforming growth factor β–activated kinase 1
- TBK1
TANK- binding kinase 1
- TGF-β1
transforming growth factor-β1
- TLR
Toll-like receptors
- TNF-α
tumor necrosis factor-α
- TRAF6
TNF receptor-associated factor
- TRIF
TIR-domain-containing adapter-inducing interferon-β
- TUDCA
tauroursodeoxycholic acid
- TXNIP
thioredoxin-interacting protein
- UPR
unfolded protein responses
- VCP
valosin-containing protein
- VDAC1
voltage-dependent anion-selective channel protein
- VSMCs
vascular smooth muscle cells
- XBP-1s
X-box-binding-protein 1 spliced isoform
Footnotes
Disclosure
The authors have no conflict of interest to declare.
References
- 1.Di Micco R, Krizhanovsky V, Baker D and d’Adda di Fagagna F. Cellular senescence in ageing: from mechanisms to therapeutic opportunities. Nat Rev Mol Cell Biol. 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Galluzzi L, Vitale I, Aaronson SA, Abrams JM, Adam D, Agostinis P, Alnemri ES, Altucci L, Amelio I, Andrews DW, et al. Molecular mechanisms of cell death: recommendations of the Nomenclature Committee on Cell Death 2018. Cell Death Differ. 2018;25:486–541. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Galluzzi L, Yamazaki T and Kroemer G. Linking cellular stress responses to systemic homeostasis. Nat Rev Mol Cell Biol. 2018;19:731–745. [DOI] [PubMed] [Google Scholar]
- 4.McCarthy CG, Wenceslau CF, Webb RC and Joe B. Novel Contributors and Mechanisms of Cellular Senescence in Hypertension-Associated Premature Vascular Aging. Am J Hypertens. 2019;32:709–719. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Dela Justina V, Giachini FR, Priviero F and Webb RC. Double-stranded RNA and Toll-like receptor activation: a novel mechanism for blood pressure regulation. Clin Sci (Lond). 2020;134:303–313. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Hypertension Cooper R., Genes, and Environment: Challenges for Prevention and Risk Prediction. Circulation. 2018;137:662–664. [DOI] [PubMed] [Google Scholar]
- 7.Cooper HA, Scalia R, Rizzo V and Eguchi S. Angiotensin II- and Alzheimer-Type Cardiovascular Aging. Circ Res. 2018;123:651–653. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Lerman LO, Kurtz TW, Touyz RM, Ellison DH, Chade AR, Crowley SD, Mattson DL, Mullins JJ, Osborn J, Eirin A, et al. Animal Models of Hypertension: A Scientific Statement From the American Heart Association. Hypertension. 2019;73:e87–e120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Martinez-Pinna R, Ramos-Mozo P, Madrigal-Matute J, Blanco-Colio LM, Lopez JA, Calvo E, Camafeita E, Lindholt JS, Meilhac O, Delbosc S, et al. Identification of peroxiredoxin-1 as a novel biomarker of abdominal aortic aneurysm. Arterioscler Thromb Vasc Biol. 2011;31:935–43. [DOI] [PubMed] [Google Scholar]
- 10.Abdellatif M, Trummer-Herbst V, Koser F, Durand S, Adão R, Vasques-Nóvoa F, Freundt JK, Voglhuber J, Pricolo MR, Kasa M, et al. Nicotinamide for the treatment of heart failure with preserved ejection fraction. Sci Transl Med. 2021;13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Chen K and Sun Z. Activation of DNA demethylases attenuates aging-associated arterial stiffening and hypertension. Aging Cell. 2018;17:e12762. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Ferrari R. RAAS inhibition and mortality in hypertension. Glob Cardiol Sci Pract. 2013;2013:269–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Nehme A and Zibara K. Efficiency and specificity of RAAS inhibitors in cardiovascular diseases: how to achieve better end-organ protection? Hypertens Res. 2017;40:903–909. [DOI] [PubMed] [Google Scholar]
- 14.Cunha PG, Boutouyrie P, Nilsson PM and Laurent S. Early Vascular Ageing (EVA): Definitions and Clinical Applicability. Curr Hypertens Rev. 2017;13:8–15. [DOI] [PubMed] [Google Scholar]
- 15.Nilsson PM. Early Vascular Aging in Hypertension. Front Cardiovasc Med. 2020;7:6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Bruno RM, Nilsson PM, Engstrom G, Wadstrom BN, Empana JP, Boutouyrie P and Laurent S. Early and Supernormal Vascular Aging: Clinical Characteristics and Association With Incident Cardiovascular Events. Hypertension. 2020;76:1616–1624. [DOI] [PubMed] [Google Scholar]
- 17.Harvey A, Montezano AC and Touyz RM. Vascular biology of ageing-Implications in hypertension. J Mol Cell Cardiol. 2015;83:112–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Camargo Livia L, Harvey Adam P, Rios Francisco J, Tsiropoulou S, Da Silva Renée de Nazaré O, Cao Z, Graham D, McMaster C, Burchmore Richard J, Hartley Richard C, et al. Vascular Nox (NADPH Oxidase) Compartmentalization, Protein Hyperoxidation, and Endoplasmic Reticulum Stress Response in Hypertension. Hypertension. 2018;72:235–246. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Kassan M, Galán M, Partyka M, Saifudeen Z, Henrion D, Trebak M and Matrougui K. Endoplasmic reticulum stress is involved in cardiac damage and vascular endothelial dysfunction in hypertensive mice. Arterioscler Thromb Vasc Biol. 2012;32:1652–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Spitler KM and Webb RC. Endoplasmic reticulum stress contributes to aortic stiffening via proapoptotic and fibrotic signaling mechanisms. Hypertension. 2014;63:e40–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Takayanagi T, Kawai T, Forrester SJ, Obama T, Tsuji T, Fukuda Y, Elliott KJ, Tilley DG, Davisson RL, Park JY, et al. Role of epidermal growth factor receptor and endoplasmic reticulum stress in vascular remodeling induced by angiotensin II. Hypertension. 2015;65:1349–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.McLellan MA, Skelly DA, Dona MSI, Squiers GT, Farrugia GE, Gaynor TL, Cohen CD, Pandey R, Diep H, Vinh A, et al. High-Resolution Transcriptomic Profiling of the Heart During Chronic Stress Reveals Cellular Drivers of Cardiac Fibrosis and Hypertrophy. Circulation. 2020;142:1448–1463. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Ayyadevara S, Mercanti F, Wang X, Mackintosh Samuel G, Tackett Alan J, Prayaga Sastry VS, Romeo F, Shmookler Reis Robert J and Mehta Jawahar L. Age- and Hypertension-Associated Protein Aggregates in Mouse Heart Have Similar Proteomic Profiles. Hypertension. 2016;67:1006–1013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Cheema MU, Poulsen ET, Enghild JJ, Hoorn EJ, Fenton RA and Praetorius J. Aldosterone and angiotensin II induce protein aggregation in renal proximal tubules. Physiol Rep. 2013;1:e00064. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Minamino T and Kitakaze M. ER stress in cardiovascular disease. J Mol Cell Cardiol. 2010;48:1105–10. [DOI] [PubMed] [Google Scholar]
- 26.Cicalese S, Okuno K and Eguchi S. Novel Methods Article Detection of protein aggregation and proteotoxicity induced by angiotensin II in vascular smooth muscle cells. J Cardiovasc Pharmacol. 2020. [DOI] [PubMed] [Google Scholar]
- 27.Kaplon RE, Chung E, Reese L, Cox-York K, Seals DR and Gentile CL. Activation of the unfolded protein response in vascular endothelial cells of nondiabetic obese adults. J Clin Endocrinol Metab. 2013;98:E1505–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Cicalese S, Okuno K, Elliott KJ, Kawai T, Scalia R, Rizzo V and Eguchi S. 78 kDa Glucose-Regulated Protein Attenuates Protein Aggregation and Monocyte Adhesion Induced by Angiotensin II in Vascular Cells. Int J Mol Sci. 2020;21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Hong J, Kim K, Kim JH and Park Y. The Role of Endoplasmic Reticulum Stress in Cardiovascular Disease and Exercise. Int J Vasc Med. 2017;2017:2049217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Hetz C, Zhang K and Kaufman RJ. Mechanisms, regulation and functions of the unfolded protein response. Nat Rev Mol Cell Biol. 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Kassan M, Ait-Aissa K, Radwan E, Mali V, Haddox S, Gabani M, Zhang W, Belmadani S, Irani K, Trebak M, et al. Essential Role of Smooth Muscle STIM1 in Hypertension and Cardiovascular Dysfunction. Arterioscler Thromb Vasc Biol. 2016;36:1900–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Mohammed-Ali Z, Lu C, Marway MK, Carlisle RE, Ask K, Lukic D, Krepinsky JC and Dickhout JG. Endoplasmic reticulum stress inhibition attenuates hypertensive chronic kidney disease through reduction in proteinuria. Sci Rep. 2017;7:41572. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Zhou N, Ma B, Stoll S, Hays TT and Qiu H. The valosin-containing protein is a novel repressor of cardiomyocyte hypertrophy induced by pressure overload. Aging Cell. 2017;16:1168–1179. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Liu TH, Tao WC, Liang QE, Tu WQ, Xiao Y and Chen LG. Gut Microbiota-Related Evidence Provides New Insights Into the Association Between Activating Transcription Factor 4 and Development of Salt-Induced Hypertension in Mice. Front Cell Dev Biol. 2020;8:585995. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Robblee MM, Kim CC, Porter Abate J, Valdearcos M, Sandlund KLM, Shenoy MK, Volmer R, Iwawaki T and Koliwad SK. Saturated Fatty Acids Engage an IRE1α-Dependent Pathway to Activate the NLRP3 Inflammasome in Myeloid Cells. Cell Rep. 2016;14:2611–2623. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Thamsen M, Ghosh R, Auyeung VC, Brumwell A, Chapman HA, Backes BJ, Perara G, Maly DJ, Sheppard D and Papa FR. Small molecule inhibition of IRE1α kinase/RNase has anti-fibrotic effects in the lung. PLoS One. 2019;14:e0209824-e0209824. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Tufanli O, Telkoparan Akillilar P, Acosta-Alvear D, Kocaturk B, Onat UI, Hamid SM, Çimen I, Walter P, Weber C and Erbay E. Targeting IRE1 with small molecules counteracts progression of atherosclerosis. Proc Natl Acad Sci U S A. 2017;114:E1395–E1404. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Prinsen JK, Kannankeril PJ, Sidorova TN, Yermalitskaya LV, Boutaud O, Zagol-Ikapitte I, Barnett JV, Murphy MB, Subati T, Stark JM, et al. Highly Reactive Isolevuglandins Promote Atrial Fibrillation Caused by Hypertension. JACC Basic Transl Sci. 2020;5:602–615. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Eguchi S, Kawai T, Scalia R and Rizzo V. Understanding Angiotensin II Type 1 Receptor Signaling in Vascular Pathophysiology. Hypertension. 2018;71:804–810. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Kawai T, Elliott KJ, Scalia R and Eguchi S. Contribution of ADAM17 and related ADAMs in cardiovascular diseases. Cellular and Molecular Life Sciences. 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Zhao W, Li Y, Jia L, Pan L, Li H and Du J. Atg5 deficiency-mediated mitophagy aggravates cardiac inflammation and injury in response to angiotensin II. Free Radic Biol Med. 2014;69:108–15. [DOI] [PubMed] [Google Scholar]
- 42.Zhang X, Li ZL, Crane JA, Jordan KL, Pawar AS, Textor SC, Lerman A and Lerman LO. Valsartan regulates myocardial autophagy and mitochondrial turnover in experimental hypertension. Hypertension. 2014;64:87–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Mondaca-Ruff D, Riquelme JA, Quiroga C, Norambuena-Soto I, Sanhueza-Olivares F, Villar-Fincheira P, Hernandez-Diaz T, Cancino-Arenas N, San Martin A, Garcia L, et al. Angiotensin II-Regulated Autophagy Is Required for Vascular Smooth Muscle Cell Hypertrophy. Front Pharmacol. 2018;9:1553. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.McCarthy CG, Wenceslau CF, Calmasini FB, Klee NS, Brands MW, Joe B and Webb RC. Reconstitution of autophagy ameliorates vascular function and arterial stiffening in spontaneously hypertensive rats. Am J Physiol Heart Circ Physiol. 2019;317:H1013–H1027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Rzymski T, Milani M, Pike L, Buffa F, Mellor HR, Winchester L, Pires I, Hammond E, Ragoussis I and Harris AL. Regulation of autophagy by ATF4 in response to severe hypoxia. Oncogene. 2010;29:4424–35. [DOI] [PubMed] [Google Scholar]
- 46.Pakos-Zebrucka K, Koryga I, Mnich K, Ljujic M, Samali A and Gorman AM. The integrated stress response. EMBO Rep. 2016;17:1374–1395. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Dikalov SI and Nazarewicz RR. Angiotensin II-induced production of mitochondrial reactive oxygen species: potential mechanisms and relevance for cardiovascular disease. Antioxid Redox Signal. 2013;19:1085–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Fukai T and Ushio-Fukai M. Superoxide dismutases: role in redox signaling, vascular function, and diseases. Antioxidants & redox signaling. 2011;15:1583–1606. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Dikalova AE, Pandey A, Xiao L, Arslanbaeva L, Sidorova T, Lopez MG, Billings FTt, Verdin E, Auwerx J, Harrison DG, et al. Mitochondrial Deacetylase Sirt3 Reduces Vascular Dysfunction and Hypertension While Sirt3 Depletion in Essential Hypertension Is Linked to Vascular Inflammation and Oxidative Stress. Circ Res. 2020;126:439–452. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.He J, Liu X, Su C, Wu F, Sun J, Zhang J, Yang X, Zhang C, Zhou Z, Zhang X, et al. Inhibition of Mitochondrial Oxidative Damage Improves Reendothelialization Capacity of Endothelial Progenitor Cells via SIRT3 (Sirtuin 3)-Enhanced SOD2 (Superoxide Dismutase 2) Deacetylation in Hypertension. Arterioscler Thromb Vasc Biol. 2019;39:1682–1698. [DOI] [PubMed] [Google Scholar]
- 51.Dikalova A, Mayorov V, Xiao L, Panov A, Amarnath V, Zagol-Ikapitte I, Vergeade A, Ao M, Yermalitsky V, Nazarewicz RR, et al. Mitochondrial Isolevuglandins Contribute to Vascular Oxidative Stress and Mitochondria-Targeted Scavenger of Isolevuglandins Reduces Mitochondrial Dysfunction and Hypertension. Hypertension. 2020:Hypertensionaha12015236. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Xiang R, Chen J, Li S, Yan H, Meng Y, Cai J, Cui Q, Yang Y, Xu M, Geng B, et al. VSMC-Specific Deletion of FAM3A Attenuated Ang II-Promoted Hypertension and Cardiovascular Hypertrophy. Circ Res. 2020;126:1746–1759. [DOI] [PubMed] [Google Scholar]
- 53.Abadir PM, Foster DB, Crow M, Cooke CA, Rucker JJ, Jain A, Smith BJ, Burks TN, Cohn RD, Fedarko NS, et al. Identification and characterization of a functional mitochondrial angiotensin system. Proc Natl Acad Sci U S A. 2011;108:14849–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Li XC, Zhou X and Zhuo JL. Evidence for a Physiological Mitochondrial Angiotensin II System in the Kidney Proximal Tubules: Novel Roles of Mitochondrial Ang II/AT(1a)/O(2)(−) and Ang II/AT(2)/NO Signaling. Hypertension. 2020;76:121–132. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Vásquez-Trincado C, García-Carvajal I, Pennanen C, Parra V, Hill JA, Rothermel BA and Lavandero S. Mitochondrial dynamics, mitophagy and cardiovascular disease. J Physiol. 2016;594:509–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Archer SL. Mitochondrial dynamics--mitochondrial fission and fusion in human diseases. N Engl J Med. 2013;369:2236–51. [DOI] [PubMed] [Google Scholar]
- 57.Killackey SA, Philpott DJ and Girardin SE. Mitophagy pathways in health and disease. J Cell Biol. 2020;219. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Gao S and Hu J. Mitochondrial Fusion: The Machineries In and Out. Trends Cell Biol. 2020. [DOI] [PubMed] [Google Scholar]
- 59.Huang G, Cong Z, Wang X, Yuan Y, Xu R, Lu Z, Wang X and Qi J. Targeting HSP90 attenuates angiotensin II-induced adventitial remodelling via suppression of mitochondrial fission. Cardiovasc Res. 2020;116:1071–1084. [DOI] [PubMed] [Google Scholar]
- 60.Cooper HA, Cicalese S, Preston KJ, Kawai T, Okuno K, Choi ET, Kasahara S, Uchida HA, Otaka N, Scalia R, et al. Targeting Mitochondrial Fission as a Potential Therapeutic for Abdominal Aortic Aneurysm. Cardiovasc Res. 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Hu Q, Zhang H, Gutiérrez Cortés N, Wu D, Wang P, Zhang J, Mattison JA, Smith E, Bettcher LF, Wang M, et al. Increased Drp1 Acetylation by Lipid Overload Induces Cardiomyocyte Death and Heart Dysfunction. Circ Res. 2020;126:456–470. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Forrester SJ, Preston KJ, Cooper HA, Boyer MJ, Escoto KM, Poltronetti AJ, Elliott KJ, Kuroda R, Miyao M, Sesaki H, et al. Mitochondrial Fission Mediates Endothelial Inflammation. Hypertension. 2020;76:267–276. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Umezu R, Koga JI, Matoba T, Katsuki S, Wang L, Hasuzawa N, Nomura M, Tsutsui H and Egashira K. Macrophage (Drp1) Dynamin-Related Protein 1 Accelerates Intimal Thickening After Vascular Injury. Arterioscler Thromb Vasc Biol. 2020;40:e214–e226. [DOI] [PubMed] [Google Scholar]
- 64.Liu X, Tan H, Liu X and Wu Q. Correlation between the expression of Drp1 in vascular endothelial cells and inflammatory factors in hypertension rats. Exp Ther Med. 2018;15:3892–3898. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Lu Y, Li S, Wu H, Bian Z, Xu J, Gu C, Chen X and Yang D. Beneficial effects of astragaloside IV against angiotensin II-induced mitochondrial dysfunction in rat vascular smooth muscle cells. Int J Mol Med. 2015;36:1223–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Swiader A, Nahapetyan H, Faccini J, D’Angelo R, Mucher E, Elbaz M, Boya P and Vindis C. Mitophagy acts as a safeguard mechanism against human vascular smooth muscle cell apoptosis induced by atherogenic lipids. Oncotarget. 2016;7:28821–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Bhatia D, Chung KP, Nakahira K, Patino E, Rice MC, Torres LK, Muthukumar T, Choi AM, Akchurin OM and Choi ME. Mitophagy-dependent macrophage reprogramming protects against kidney fibrosis. JCI Insight. 2019;4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Motwani M, Pesiridis S and Fitzgerald KA. DNA sensing by the cGAS-STING pathway in health and disease. Nat Rev Genet. 2019;20:657–674. [DOI] [PubMed] [Google Scholar]
- 69.Liu H, Cheng WL, Jiang X, Wang PX, Fang C, Zhu XY, Huang Z, She ZG and Li H. Ablation of Interferon Regulatory Factor 3 Protects Against Atherosclerosis in Apolipoprotein E-Deficient Mice. Hypertension. 2017;69:510–520. [DOI] [PubMed] [Google Scholar]
- 70.Tsushima K, Osawa T, Yanai H, Nakajima A, Takaoka A, Manabe I, Ohba Y, Imai Y, Taniguchi T and Nagai R. IRF3 regulates cardiac fibrosis but not hypertrophy in mice during angiotensin II-induced hypertension. FASEB J. 2011;25:1531–43. [DOI] [PubMed] [Google Scholar]
- 71.Mao Y, Luo W, Zhang L, Wu W, Yuan L, Xu H, Song J, Fujiwara K, Abe JI, LeMaire SA, et al. STING-IRF3 Triggers Endothelial Inflammation in Response to Free Fatty Acid-Induced Mitochondrial Damage in Diet-Induced Obesity. Arterioscler Thromb Vasc Biol. 2017;37:920–929. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Pacher P, Sharma K, Csordas G, Zhu Y and Hajnoczky G. Uncoupling of ER-mitochondrial calcium communication by transforming growth factor-beta. Am J Physiol Renal Physiol. 2008;295:F1303–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Adebiyi A, Thomas-Gatewood CM, Leo MD, Kidd MW, Neeb ZP and Jaggar JH. An elevation in physical coupling of type 1 inositol 1,4,5-trisphosphate (IP3) receptors to transient receptor potential 3 (TRPC3) channels constricts mesenteric arteries in genetic hypertension. Hypertension. 2012;60:1213–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Matthews C, Gorenne I, Scott S, Figg N, Kirkpatrick P, Ritchie A, Goddard M and Bennett M. Vascular smooth muscle cells undergo telomere-based senescence in human atherosclerosis: effects of telomerase and oxidative stress. Circ Res. 2006;99:156–64. [DOI] [PubMed] [Google Scholar]
- 75.Childs BG, Baker DJ, Wijshake T, Conover CA, Campisi J and van Deursen JM. Senescent intimal foam cells are deleterious at all stages of atherosclerosis. Science. 2016;354:472–477. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Walaszczyk A, Dookun E, Redgrave R, Tual-Chalot S, Victorelli S, Spyridopoulos I, Owens A, Arthur HM, Passos JF and Richardson GD. Pharmacological clearance of senescent cells improves survival and recovery in aged mice following acute myocardial infarction. Aging Cell. 2019;18:e12945. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Novella S, Dantas AP, Segarra G, Vidal-Gomez X, Mompeon A, Garabito M, Hermenegildo C and Medina P. Aging-related endothelial dysfunction in the aorta from female senescence-accelerated mice is associated with decreased nitric oxide synthase expression. Exp Gerontol. 2013;48:1329–37. [DOI] [PubMed] [Google Scholar]
- 78.Kunieda T, Minamino T, Nishi J, Tateno K, Oyama T, Katsuno T, Miyauchi H, Orimo M, Okada S, Takamura M, et al. Angiotensin II induces premature senescence of vascular smooth muscle cells and accelerates the development of atherosclerosis via a p21-dependent pathway. Circulation. 2006;114:953–60. [DOI] [PubMed] [Google Scholar]
- 79.Li DJ, Huang F, Ni M, Fu H, Zhang LS and Shen FM. alpha7 Nicotinic Acetylcholine Receptor Relieves Angiotensin II-Induced Senescence in Vascular Smooth Muscle Cells by Raising Nicotinamide Adenine Dinucleotide-Dependent SIRT1 Activity. Arterioscler Thromb Vasc Biol. 2016;36:1566–76. [DOI] [PubMed] [Google Scholar]
- 80.Xu XJ, Zhao WB, Feng SB, Sun C, Chen Q, Ni B and Hu HY. Celastrol alleviates angiotensin IImediated vascular smooth muscle cell senescence via induction of autophagy. Mol Med Rep. 2017;16:7657–7664. [DOI] [PubMed] [Google Scholar]
- 81.Okuno K, Cicalese S, Elliott KJ, Kawai T, Hashimoto T and Eguchi S. Targeting Molecular Mechanism of Vascular Smooth Muscle Senescence Induced by Angiotensin II, A Potential Therapy via Senolytics and Senomorphics. Int J Mol Sci. 2020;21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Tsai IC, Pan ZC, Cheng HP, Liu CH, Lin BT and Jiang MJ. Reactive oxygen species derived from NADPH oxidase 1 and mitochondria mediate angiotensin II-induced smooth muscle cell senescence. J Mol Cell Cardiol. 2016;98:18–27. [DOI] [PubMed] [Google Scholar]
- 83.Chen HZ, Wang F, Gao P, Pei JF, Liu Y, Xu TT, Tang X, Fu WY, Lu J, Yan YF, et al. Age-Associated Sirtuin 1 Reduction in Vascular Smooth Muscle Links Vascular Senescence and Inflammation to Abdominal Aortic Aneurysm. Circ Res. 2016;119:1076–1088. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Imanishi T, Moriwaki C, Hano T and Nishio I. Endothelial progenitor cell senescence is accelerated in both experimental hypertensive rats and patients with essential hypertension. J Hypertens. 2005;23:1831–7. [DOI] [PubMed] [Google Scholar]
- 85.Westhoff JH, Hilgers KF, Steinbach MP, Hartner A, Klanke B, Amann K and Melk A. Hypertension induces somatic cellular senescence in rats and humans by induction of cell cycle inhibitor p16INK4a. Hypertension. 2008;52:123–9. [DOI] [PubMed] [Google Scholar]
- 86.Morgan RG, Ives SJ, Walker AE, Cawthon RM, Andtbacka RH, Noyes D, Lesniewski LA, Richardson RS and Donato AJ. Role of arterial telomere dysfunction in hypertension: relative contributions of telomere shortening and telomere uncapping. J Hypertens. 2014;32:1293–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Hernandez-Segura A, Nehme J and Demaria M. Hallmarks of Cellular Senescence. Trends Cell Biol. 2018;28:436–453. [DOI] [PubMed] [Google Scholar]
- 88.McHugh D and Gil J. Senescence and aging: Causes, consequences, and therapeutic avenues. J Cell Biol. 2018;217:65–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Coppe JP, Desprez PY, Krtolica A and Campisi J. The senescence-associated secretory phenotype: the dark side of tumor suppression. Annu Rev Pathol. 2010;5:99–118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Franceschi C and Campisi J. Chronic inflammation (inflammaging) and its potential contribution to age-associated diseases. J Gerontol A Biol Sci Med Sci. 2014;69 Suppl 1:S4–9. [DOI] [PubMed] [Google Scholar]
- 91.Roos CM, Zhang B, Palmer AK, Ogrodnik MB, Pirtskhalava T, Thalji NM, Hagler M, Jurk D, Smith LA, Casaclang-Verzosa G, et al. Chronic senolytic treatment alleviates established vasomotor dysfunction in aged or atherosclerotic mice. Aging Cell. 2016;15:973–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Miyao M, Cicalese S, Kawai T, Cooper HA, Boyer MJ, Elliott KJ, Forrester SJ, Kuroda R, Rizzo V, Hashimoto T, et al. Involvement of Senescence and Mitochondrial Fission in Endothelial Cell Pro-Inflammatory Phenotype Induced by Angiotensin II. Int J Mol Sci. 2020;21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Taylor RC and Hetz C. Mastering organismal aging through the endoplasmic reticulum proteostasis network. Aging Cell. 2020;19:e13265. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Abbadie C and Pluquet O. Unfolded Protein Response (UPR) Controls Major Senescence Hallmarks. Trends Biochem Sci. 2020;45:371–374. [DOI] [PubMed] [Google Scholar]
- 95.Hamczyk MR, Villa-Bellosta R, Quesada V, Gonzalo P, Vidak S, Nevado RM, Andres-Manzano MJ, Misteli T, Lopez-Otin C and Andres V. Progerin accelerates atherosclerosis by inducing endoplasmic reticulum stress in vascular smooth muscle cells. EMBO Mol Med. 2019;11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Biwer LA, Askew-Page HR, Hong K, Milstein J, Johnstone SR, Macal E, Good ME, Bagher P, Sonkusare SK and Isakson BE. Endothelial calreticulin deletion impairs endothelial function in aged mice. Am J Physiol Heart Circ Physiol. 2020;318:H1041–H1048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Lee HY, Kim HK, Hoang TH, Yang S, Kim HR and Chae HJ. The correlation of IRE1alpha oxidation with Nox4 activation in aging-associated vascular dysfunction. Redox Biol. 2020;37:101727. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Rossman MJ, Gioscia-Ryan RA, Clayton ZS, Murphy MP and Seals DR. Targeting mitochondrial fitness as a strategy for healthy vascular aging. Clin Sci (Lond). 2020;134:1491–1519. [DOI] [PubMed] [Google Scholar]
- 99.Rossman MJ, Santos-Parker JR, Steward CAC, Bispham NZ, Cuevas LM, Rosenberg HL, Woodward KA, Chonchol M, Gioscia-Ryan RA, Murphy MP, et al. Chronic Supplementation With a Mitochondrial Antioxidant (MitoQ) Improves Vascular Function in Healthy Older Adults. Hypertension. 2018;71:1056–1063. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Sural S, Liang CY, Wang FY, Ching TT and Hsu AL. HSB-1/HSF-1 pathway modulates histone H4 in mitochondria to control mtDNA transcription and longevity. Sci Adv. 2020;6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Lin JR, Shen WL, Yan C and Gao PJ. Downregulation of dynamin-related protein 1 contributes to impaired autophagic flux and angiogenic function in senescent endothelial cells. Arterioscler Thromb Vasc Biol. 2015;35:1413–22. [DOI] [PubMed] [Google Scholar]
- 102.Costa TJ, Barros PR, Arce C, Santos JD, da Silva-Neto J, Egea G, Dantas AP, Tostes RC and Jimenez-Altayo F. The homeostatic role of hydrogen peroxide, superoxide anion and nitric oxide in the vasculature. Free Radic Biol Med. 2020. [DOI] [PubMed] [Google Scholar]
- 103.Incalza MA, D’Oria R, Natalicchio A, Perrini S, Laviola L and Giorgino F. Oxidative stress and reactive oxygen species in endothelial dysfunction associated with cardiovascular and metabolic diseases. Vascul Pharmacol. 2018;100:1–19. [DOI] [PubMed] [Google Scholar]
- 104.Ferreira NS, Tostes RC, Paradis P and Schiffrin EL. Aldosterone, Inflammation, Immune System and Hypertension. Am J Hypertens. 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Guzik TJ and Touyz RM. Oxidative Stress, Inflammation, and Vascular Aging in Hypertension. Hypertension. 2017;70:660–667. [DOI] [PubMed] [Google Scholar]
- 106.Reckelhoff JF, Romero DG and Yanes Cardozo LL. Sex, Oxidative Stress, and Hypertension: Insights From Animal Models. Physiology (Bethesda). 2019;34:178–188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Papaconstantinou J. The Role of Signaling Pathways of Inflammation and Oxidative Stress in Development of Senescence and Aging Phenotypes in Cardiovascular Disease. Cells. 2019;8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Yamamoto M, Kensler TW and Motohashi H. The KEAP1-NRF2 System: a Thiol-Based Sensor-Effector Apparatus for Maintaining Redox Homeostasis. Physiol Rev. 2018;98:1169–1203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Itoh K, Chiba T, Takahashi S, Ishii T, Igarashi K, Katoh Y, Oyake T, Hayashi N, Satoh K, Hatayama I, et al. An Nrf2/small Maf heterodimer mediates the induction of phase II detoxifying enzyme genes through antioxidant response elements. Biochem Biophys Res Commun. 1997;236:313–22. [DOI] [PubMed] [Google Scholar]
- 110.Zheng X, Wang G, Bin P, Meng T, Niu Y, Yang M, Zhang L, Duan H, Yu T, Dai Y, et al. Time-course effects of antioxidants and phase II enzymes on diesel exhaust particles-induced oxidative damage in the mouse lung. Toxicol Appl Pharmacol. 2019;366:25–34. [DOI] [PubMed] [Google Scholar]
- 111.Favreau LV and Pickett CB. Transcriptional regulation of the rat NAD(P)H:quinone reductase gene. Characterization of a DNA-protein interaction at the antioxidant responsive element and induction by 12-O-tetradecanoylphorbol 13-acetate. J Biol Chem. 1993;268:19875–81. [PubMed] [Google Scholar]
- 112.Rushmore TH, Morton MR and Pickett CB. The antioxidant responsive element. Activation by oxidative stress and identification of the DNA consensus sequence required for functional activity. J Biol Chem. 1991;266:11632–9. [PubMed] [Google Scholar]
- 113.Itoh K, Wakabayashi N, Katoh Y, Ishii T, Igarashi K, Engel JD and Yamamoto M. Keap1 represses nuclear activation of antioxidant responsive elements by Nrf2 through binding to the amino-terminal Neh2 domain. Genes Dev. 1999;13:76–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Cullinan SB, Gordan JD, Jin J, Harper JW and Diehl JA. The Keap1-BTB protein is an adaptor that bridges Nrf2 to a Cul3-based E3 ligase: oxidative stress sensing by a Cul3-Keap1 ligase. Mol Cell Biol. 2004;24:8477–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Zhang DD, Lo SC, Cross JV, Templeton DJ and Hannink M. Keap1 is a redox-regulated substrate adaptor protein for a Cul3-dependent ubiquitin ligase complex. Mol Cell Biol. 2004;24:10941–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Wang C, Luo Z, Carter G, Wellstein A, Jose PA, Tomlinson J, Leiper J, Welch WJ, Wilcox CS and Wang D. NRF2 prevents hypertension, increased ADMA, microvascular oxidative stress, and dysfunction in mice with two weeks of ANG II infusion. Am J Physiol Regul Integr Comp Physiol. 2018;314:R399–R406. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Lopes RA, Neves KB, Tostes RC, Montezano AC and Touyz RM. Downregulation of Nuclear Factor Erythroid 2-Related Factor and Associated Antioxidant Genes Contributes to Redox-Sensitive Vascular Dysfunction in Hypertension. Hypertension. 2015;66:1240–50. [DOI] [PubMed] [Google Scholar]
- 118.Zakkar M, Van der Heiden K, Luong le A, Chaudhury H, Cuhlmann S, Hamdulay SS, Krams R, Edirisinghe I, Rahman I, Carlsen H, et al. Activation of Nrf2 in endothelial cells protects arteries from exhibiting a proinflammatory state. Arterioscler Thromb Vasc Biol. 2009;29:1851–7. [DOI] [PubMed] [Google Scholar]
- 119.Florczyk U, Jazwa A, Maleszewska M, Mendel M, Szade K, Kozakowska M, Grochot-Przeczek A, Viscardi M, Czauderna S, Bukowska-Strakova K, et al. Nrf2 regulates angiogenesis: effect on endothelial cells, bone marrow-derived proangiogenic cells and hind limb ischemia. Antioxid Redox Signal. 2014;20:1693–708. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Amaral JH, Rizzi ES, Alves-Lopes R, Pinheiro LC, Tostes RC and Tanus-Santos JE. Antioxidant and antihypertensive responses to oral nitrite involves activation of the Nrf2 pathway. Free Radic Biol Med. 2019;141:261–268. [DOI] [PubMed] [Google Scholar]
- 121.Gao L, Zimmerman MC, Biswal S and Zucker IH. Selective Nrf2 Gene Deletion in the Rostral Ventrolateral Medulla Evokes Hypertension and Sympathoexcitation in Mice. Hypertension. 2017;69:1198–1206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Shimoyama Y, Mitsuda Y, Hamajima N and Niwa T. Polymorphisms of Nrf2, an antioxidative gene, are associated with blood pressure in Japanese. Nagoya J Med Sci. 2014;76:113–20. [PMC free article] [PubMed] [Google Scholar]
- 123.Testa A, Leonardis D, Spoto B, Sanguedolce MC, Parlongo RM, Pisano A, Tripepi G, Mallamaci F and Zoccali C. A polymorphism in a major antioxidant gene (Kelch-like ECH-associated protein 1) predicts incident cardiovascular events in chronic kidney disease patients: an exploratory study. J Hypertens. 2016;34:928–34. [DOI] [PubMed] [Google Scholar]
- 124.Li J, Zhang C, Xing Y, Janicki JS, Yamamoto M, Wang XL, Tang DQ and Cui T. Up-regulation of p27(kip1) contributes to Nrf2-mediated protection against angiotensin II-induced cardiac hypertrophy. Cardiovasc Res. 2011;90:315–24. [DOI] [PubMed] [Google Scholar]
- 125.Rush BM, Bondi CD, Stocker SD, Barry KM, Small SA, Ong J, Jobbagy S, Stolz DB, Bastacky SI, Chartoumpekis DV, et al. Genetic or pharmacologic Nrf2 activation increases proteinuria in chronic kidney disease in mice. Kidney Int. 2021;99:102–116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Neves KB, Montezano AC, Alves-Lopes R, Bruder-Nascimento T, Costa RM, Costa RS, Touyz RM and Tostes RC. Upregulation of Nrf2 and Decreased Redox Signaling Contribute to Renoprotective Effects of Chemerin Receptor Blockade in Diabetic Mice. Int J Mol Sci. 2018;19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Neves KB, Nguyen Dinh Cat A, Alves-Lopes R, Harvey KY, Costa RMD, Lobato NS, Montezano AC, Oliveira AM, Touyz RM and Tostes RC. Chemerin receptor blockade improves vascular function in diabetic obese mice via redox-sensitive and Akt-dependent pathways. Am J Physiol Heart Circ Physiol. 2018;315:H1851–H1860. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Li MH, Cha YN and Surh YJ. Peroxynitrite induces HO-1 expression via PI3K/Akt-dependent activation of NF-E2-related factor 2 in PC12 cells. Free Radic Biol Med. 2006;41:1079–91. [DOI] [PubMed] [Google Scholar]
- 129.Salazar M, Rojo AI, Velasco D, de Sagarra RM and Cuadrado A. Glycogen synthase kinase-3beta inhibits the xenobiotic and antioxidant cell response by direct phosphorylation and nuclear exclusion of the transcription factor Nrf2. J Biol Chem. 2006;281:14841–51. [DOI] [PubMed] [Google Scholar]
- 130.Rada P, Rojo AI, Evrard-Todeschi N, Innamorato NG, Cotte A, Jaworski T, Tobon-Velasco JC, Devijver H, Garcia-Mayoral MF, Van Leuven F, et al. Structural and functional characterization of Nrf2 degradation by the glycogen synthase kinase 3/beta-TrCP axis. Mol Cell Biol. 2012;32:3486–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Alves-Lopes R, Neves KB, Montezano AC, Harvey A, Carneiro FS, Touyz RM and Tostes RC. Internal Pudental Artery Dysfunction in Diabetes Mellitus Is Mediated by NOX1-Derived ROS-, Nrf2-, and Rho Kinase-Dependent Mechanisms. Hypertension. 2016;68:1056–64. [DOI] [PubMed] [Google Scholar]
- 132.Kobayashi EH, Suzuki T, Funayama R, Nagashima T, Hayashi M, Sekine H, Tanaka N, Moriguchi T, Motohashi H, Nakayama K, et al. Nrf2 suppresses macrophage inflammatory response by blocking proinflammatory cytokine transcription. Nat Commun. 2016;7:11624. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Hayes JD and Dinkova-Kostova AT. The Nrf2 regulatory network provides an interface between redox and intermediary metabolism. Trends Biochem Sci. 2014;39:199–218. [DOI] [PubMed] [Google Scholar]
- 134.Chapple SJ, Siow RC and Mann GE. Crosstalk between Nrf2 and the proteasome: therapeutic potential of Nrf2 inducers in vascular disease and aging. Int J Biochem Cell Biol. 2012;44:1315–20. [DOI] [PubMed] [Google Scholar]
- 135.Kim KM, Pae HO, Zheng M, Park R, Kim YM and Chung HT. Carbon monoxide induces heme oxygenase-1 via activation of protein kinase R-like endoplasmic reticulum kinase and inhibits endothelial cell apoptosis triggered by endoplasmic reticulum stress. Circ Res. 2007;101:919–27. [DOI] [PubMed] [Google Scholar]
- 136.Deng S, Essandoh K, Wang X, Li Y, Huang W, Chen J, Peng J, Jiang DS, Mu X, Wang C, et al. Tsg101 positively regulates P62-Keap1-Nrf2 pathway to protect hearts against oxidative damage. Redox Biol. 2020;32:101453. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Ungvari Z, Bailey-Downs L, Sosnowska D, Gautam T, Koncz P, Losonczy G, Ballabh P, de Cabo R, Sonntag WE and Csiszar A. Vascular oxidative stress in aging: a homeostatic failure due to dysregulation of NRF2-mediated antioxidant response. Am J Physiol Heart Circ Physiol. 2011;301:H363–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Ungvari Z, Bailey-Downs L, Gautam T, Sosnowska D, Wang M, Monticone RE, Telljohann R, Pinto JT, de Cabo R, Sonntag WE, et al. Age-associated vascular oxidative stress, Nrf2 dysfunction, and NF-{kappa}B activation in the nonhuman primate Macaca mulatta. J Gerontol A Biol Sci Med Sci. 2011;66:866–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Fulop GA, Kiss T, Tarantini S, Balasubramanian P, Yabluchanskiy A, Farkas E, Bari F, Ungvari Z and Csiszar A. Nrf2 deficiency in aged mice exacerbates cellular senescence promoting cerebrovascular inflammation. Geroscience. 2018;40:513–521. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Kuosmanen SM, Sihvola V, Kansanen E, Kaikkonen MU and Levonen AL. MicroRNAs mediate the senescence-associated decline of NRF2 in endothelial cells. Redox Biol. 2018;18:77–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Kloska D, Kopacz A, Piechota-Polanczyk A, Nowak WN, Dulak J, Jozkowicz A and Grochot-Przeczek A. Nrf2 in aging - Focus on the cardiovascular system. Vascul Pharmacol. 2019;112:42–53. [DOI] [PubMed] [Google Scholar]
- 142.Pergola PE, Raskin P, Toto RD, Meyer CJ, Huff JW, Grossman EB, Krauth M, Ruiz S, Audhya P, Christ-Schmidt H, et al. Bardoxolone methyl and kidney function in CKD with type 2 diabetes. N Engl J Med. 2011;365:327–36. [DOI] [PubMed] [Google Scholar]
- 143.Axelsson AS, Tubbs E, Mecham B, Chacko S, Nenonen HA, Tang Y, Fahey JW, Derry JMJ, Wollheim CB, Wierup N, et al. Sulforaphane reduces hepatic glucose production and improves glucose control in patients with type 2 diabetes. Sci Transl Med. 2017;9. [DOI] [PubMed] [Google Scholar]
- 144.Arefin S, Buchanan S, Hobson S, Steinmetz J, Alsalhi S, Shiels PG, Kublickiene K and Stenvinkel P. Nrf2 in early vascular ageing: Calcification, senescence and therapy. Clin Chim Acta. 2020;505:108–118. [DOI] [PubMed] [Google Scholar]
- 145.de Zeeuw D, Akizawa T, Audhya P, Bakris GL, Chin M, Christ-Schmidt H, Goldsberry A, Houser M, Krauth M, Lambers Heerspink HJ, et al. Bardoxolone methyl in type 2 diabetes and stage 4 chronic kidney disease. N Engl J Med. 2013;369:2492–503. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Egea J, Fabregat I, Frapart YM, Ghezzi P, Gorlach A, Kietzmann T, Kubaichuk K, Knaus UG, Lopez MG, Olaso-Gonzalez G, et al. European contribution to the study of ROS: A summary of the findings and prospects for the future from the COST action BM1203 (EU-ROS). Redox Biol. 2017;13:94–162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Zhong Z, Liang S, Sanchez-Lopez E, He F, Shalapour S, Lin XJ, Wong J, Ding S, Seki E, Schnabl B, et al. New mitochondrial DNA synthesis enables NLRP3 inflammasome activation. Nature. 2018;560:198–203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Kim YG, Kim SM, Kim KP, Lee SH and Moon JY. The Role of Inflammasome-Dependent and Inflammasome-Independent NLRP3 in the Kidney. Cells. 2019;8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Kelley N, Jeltema D, Duan Y and He Y. The NLRP3 Inflammasome: An Overview of Mechanisms of Activation and Regulation. Int J Mol Sci. 2019;20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Amarante-Mendes GP, Adjemian S, Branco LM, Zanetti LC, Weinlich R and Bortoluci KR. Pattern Recognition Receptors and the Host Cell Death Molecular Machinery. Front Immunol. 2018;9:2379. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Ishihara M, Tono Y, Miyahara Y, Muraoka D, Harada N, Kageyama S, Sasaki T, Hori Y, Soga N, Uchida K, et al. First-in-human phase I clinical trial of the NY-ESO-1 protein cancer vaccine with NOD2 and TLR9 stimulants in patients with NY-ESO-1-expressing refractory solid tumors. Cancer Immunol Immunother. 2020;69:663–675. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.O’Neill LA, Golenbock D and Bowie AG. The history of Toll-like receptors - redefining innate immunity. Nat Rev Immunol. 2013;13:453–60. [DOI] [PubMed] [Google Scholar]
- 153.Mitchell JA, Ryffel B, Quesniaux VF, Cartwright N and Paul-Clark M. Role of pattern-recognition receptors in cardiovascular health and disease. Biochem Soc Trans. 2007;35:1449–52. [DOI] [PubMed] [Google Scholar]
- 154.Goulopoulou S, McCarthy CG and Webb RC. Toll-like Receptors in the Vascular System: Sensing the Dangers Within. Pharmacol Rev. 2016;68:142–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Kawasaki T and Kawai T. Toll-like receptor signaling pathways. Front Immunol. 2014;5:461. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Andrade WA, Souza Mdo C, Ramos-Martinez E, Nagpal K, Dutra MS, Melo MB, Bartholomeu DC, Ghosh S, Golenbock DT and Gazzinelli RT. Combined action of nucleic acid-sensing Toll-like receptors and TLR11/TLR12 heterodimers imparts resistance to Toxoplasma gondii in mice. Cell Host Microbe. 2013;13:42–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Oldenburg M, Kruger A, Ferstl R, Kaufmann A, Nees G, Sigmund A, Bathke B, Lauterbach H, Suter M, Dreher S, et al. TLR13 recognizes bacterial 23S rRNA devoid of erythromycin resistance-forming modification. Science. 2012;337:1111–5. [DOI] [PubMed] [Google Scholar]
- 158.Cartwright N, McMaster SK, Sorrentino R, Paul-Clark M, Sriskandan S, Ryffel B, Quesniaux VF, Evans TW and Mitchell JA. Elucidation of toll-like receptor and adapter protein signaling in vascular dysfunction induced by gram-positive Staphylococcus aureus or gram-negative Escherichia coli. Shock. 2007;27:40–7. [DOI] [PubMed] [Google Scholar]
- 159.Lin K, Luo W, Yan J, Shen S, Shen Q, Wang J, Guan X, Wu G, Huang W and Liang G. TLR2 regulates angiotensin II-induced vascular remodeling and EndMT through NF-kappaB signaling. Aging (Albany NY). 2020;12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Speer T, Rohrer L, Blyszczuk P, Shroff R, Kuschnerus K, Krankel N, Kania G, Zewinger S, Akhmedov A, Shi Y, et al. Abnormal high-density lipoprotein induces endothelial dysfunction via activation of Toll-like receptor-2. Immunity. 2013;38:754–68. [DOI] [PubMed] [Google Scholar]
- 161.Schuchardt M, Prufer N, Tu Y, Herrmann J, Hu XP, Chebli S, Dahlke K, Zidek W, van der Giet M and Tolle M. Dysfunctional high-density lipoprotein activates toll-like receptors via serum amyloid A in vascular smooth muscle cells. Sci Rep. 2019;9:3421. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.de Oliveira AA, Faustino J, Webb RC and Nunes KP. Blockade of the TLR4-MD2 complex lowers blood pressure and improves vascular function in a murine model of type 1 diabetes. Sci Rep. 2020;10:12032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Strela FB, Brun BF, Berger RCM, Melo S, de Oliveira EM, Barauna VG and Vassallo PF. Lipopolysaccharide exposure modulates the contractile and migratory phenotypes of vascular smooth muscle cells. Life Sci. 2020;241:117098. [DOI] [PubMed] [Google Scholar]
- 164.Akira S and Takeda K. Toll-like receptor signalling. Nat Rev Immunol. 2004;4:499–511. [DOI] [PubMed] [Google Scholar]
- 165.Zuo ZT, Ma Y, Sun Y, Bai CQ, Zhou HY and Chen BH. Role of TLR4/NF-kappaB Signalling Pathway in Pulmonary Arterial Hypertension in Patients with Chronic Obstructive Pulmonary Disease. J Coll Physicians Surg Pak. 2020;30:568–573. [DOI] [PubMed] [Google Scholar]
- 166.Sha H, Ma Y, Tong Y, Zhao J and Qin F. Apocynin inhibits placental TLR4/NF-kappaB signaling pathway and ameliorates preeclampsia-like symptoms in rats. Pregnancy Hypertens. 2020;22:210–215. [DOI] [PubMed] [Google Scholar]
- 167.Nunes KP, Bomfim GF, Toque HA, Szasz T and Clinton Webb R. Toll-like receptor 4 (TLR4) impairs nitric oxide contributing to Angiotensin II-induced cavernosal dysfunction. Life Sci. 2017;191:219–226. [DOI] [PubMed] [Google Scholar]
- 168.Dange RB, Agarwal D, Teruyama R and Francis J. Toll-like receptor 4 inhibition within the paraventricular nucleus attenuates blood pressure and inflammatory response in a genetic model of hypertension. J Neuroinflammation. 2015;12:31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Qi J, Yu XJ, Fu LY, Liu KL, Gao TT, Tu JW, Kang KB, Shi XL, Li HB, Li Y, et al. Exercise Training Attenuates Hypertension Through TLR4/MyD88/NF-kappaB Signaling in the Hypothalamic Paraventricular Nucleus. Front Neurosci. 2019;13:1138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Kim HY, Hong MH, Yoon JJ, Kim DS, Na SW, Jang YJ, Lee YJ, Kang DG and Lee HS. Protective Effect of Vitis labrusca Leaves Extract on Cardiovascular Dysfunction through HMGB1-TLR4-NFkappaB Signaling in Spontaneously Hypertensive Rats. Nutrients. 2020;12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Bomfim GF, Dos Santos RA, Oliveira MA, Giachini FR, Akamine EH, Tostes RC, Fortes ZB, Webb RC and Carvalho MH. Toll-like receptor 4 contributes to blood pressure regulation and vascular contraction in spontaneously hypertensive rats. Clin Sci (Lond). 2012;122:535–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Oozawa S, Sano S and Nishibori M. Usefulness of high mobility group box 1 protein as a plasma biomarker in patient with peripheral artery disease. Acta Med Okayama. 2014;68:157–62. [DOI] [PubMed] [Google Scholar]
- 173.Wang HH, Lin M and Xiang GD. Serum HMGB1 levels and its association with endothelial dysfunction in patients with polycystic ovary syndrome. Physiol Res. 2018;67:911–919. [DOI] [PubMed] [Google Scholar]
- 174.Singh MV, Cicha MZ, Nunez S, Meyerholz DK, Chapleau MW and Abboud FM. Angiotensin II-induced hypertension and cardiac hypertrophy are differentially mediated by TLR3- and TLR4-dependent pathways. Am J Physiol Heart Circ Physiol. 2019;316:H1027–H1038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Hato T and Dagher PC. How the Innate Immune System Senses Trouble and Causes Trouble. Clin J Am Soc Nephrol. 2015;10:1459–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Zimmer S, Steinmetz M, Asdonk T, Motz I, Coch C, Hartmann E, Barchet W, Wassmann S, Hartmann G and Nickenig G. Activation of endothelial toll-like receptor 3 impairs endothelial function. Circ Res. 2011;108:1358–66. [DOI] [PubMed] [Google Scholar]
- 177.Chatterjee P, Chiasson VL, Seerangan G, Tobin RP, Kopriva SE, Newell-Rogers MK and Mitchell BM. Cotreatment with interleukin 4 and interleukin 10 modulates immune cells and prevents hypertension in pregnant mice. Am J Hypertens. 2015;28:135–42. [DOI] [PubMed] [Google Scholar]
- 178.Koch SR, Choi H, Mace EH and Stark RJ. Toll-like receptor 3-mediated inflammation by p38 is enhanced by endothelial nitric oxide synthase knockdown. Cell Commun Signal. 2019;17:33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.McCarthy CG, Wenceslau CF, Goulopoulou S, Ogbi S, Baban B, Sullivan JC, Matsumoto T and Webb RC. Circulating mitochondrial DNA and Toll-like receptor 9 are associated with vascular dysfunction in spontaneously hypertensive rats. Cardiovasc Res. 2015;107:119–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.McCarthy CG, Wenceslau CF, Goulopoulou S, Baban B, Matsumoto T and Webb RC. Chloroquine Suppresses the Development of Hypertension in Spontaneously Hypertensive Rats. Am J Hypertens. 2017;30:173–181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Echem C, Costa TJD, Oliveira V, Giglio Colli L, Landgraf MA, Rodrigues SF, Franco M, Landgraf RG, Santos-Eichler RA, Bomfim GF, et al. Mitochondrial DNA: A new driver for sex differences in spontaneous hypertension. Pharmacol Res. 2019;144:142–150. [DOI] [PubMed] [Google Scholar]
- 182.Alvarado-Vasquez N. Circulating cell-free mitochondrial DNA as the probable inducer of early endothelial dysfunction in the prediabetic patient. Exp Gerontol. 2015;69:70–8. [DOI] [PubMed] [Google Scholar]
- 183.Goulopoulou S, Matsumoto T, Bomfim GF and Webb RC. Toll-like receptor 9 activation: a novel mechanism linking placenta-derived mitochondrial DNA and vascular dysfunction in pre-eclampsia. Clin Sci (Lond). 2012;123:429–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Krogmann AO, Lusebrink E, Steinmetz M, Asdonk T, Lahrmann C, Lutjohann D, Nickenig G and Zimmer S. Proinflammatory Stimulation of Toll-Like Receptor 9 with High Dose CpG ODN 1826 Impairs Endothelial Regeneration and Promotes Atherosclerosis in Mice. PLoS One. 2016;11:e0146326. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Kawai T and Akira S. Toll-like receptors and their crosstalk with other innate receptors in infection and immunity. Immunity. 2011;34:637–50. [DOI] [PubMed] [Google Scholar]
- 186.Robles-Vera I, Visitacion N, Toral M, Sanchez M, Gomez-Guzman M, O’Valle F, Jimenez R, Duarte J and Romero M. Toll-like receptor 7-driven lupus autoimmunity induces hypertension and vascular alterations in mice. J Hypertens. 2020;38:1322–1335. [DOI] [PubMed] [Google Scholar]
- 187.Chatterjee P, Weaver LE, Doersch KM, Kopriva SE, Chiasson VL, Allen SJ, Narayanan AM, Young KJ, Jones KA, Kuehl TJ, et al. Placental Toll-like receptor 3 and Toll-like receptor 7/8 activation contributes to preeclampsia in humans and mice. PLoS One. 2012;7:e41884. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Kapelouzou A, Giaglis S, Peroulis M, Katsimpoulas M, Moustardas P, Aravanis CV, Kostakis A, Karayannakos PE and Cokkinos DV. Overexpression of Toll-Like Receptors 2, 3, 4, and 8 Is Correlated to the Vascular Atherosclerotic Process in the Hyperlipidemic Rabbit Model: The Effect of Statin Treatment. J Vasc Res. 2017;54:156–169. [DOI] [PubMed] [Google Scholar]
- 189.Nunes KP and Webb RC. New insights into RhoA/Rho-kinase signaling: a key regulator of vascular contraction. Small GTPases. 2020:1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Stallmann-Jorgensen I, Ogbi S, Szasz T and Webb RC. A Toll-Like Receptor 1/2 Agonist Augments Contractility in Rat Corpus Cavernosum. J Sex Med. 2015;12:1722–31. [DOI] [PubMed] [Google Scholar]
- 191.McCarthy CG, Wenceslau CF, Ogbi S, Szasz T and Webb RC. Toll-Like Receptor 9-Dependent AMPKalpha Activation Occurs via TAK1 and Contributes to RhoA/ROCK Signaling and Actin Polymerization in Vascular Smooth Muscle Cells. J Pharmacol Exp Ther. 2018;365:60–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Krishnan SM, Ling YH, Huuskes BM, Ferens DM, Saini N, Chan CT, Diep H, Kett MM, Samuel CS, Kemp-Harper BK, et al. Pharmacological inhibition of the NLRP3 inflammasome reduces blood pressure, renal damage, and dysfunction in salt-sensitive hypertension. Cardiovasc Res. 2019;115:776–787. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Sun HJ, Ren XS, Xiong XQ, Chen YZ, Zhao MX, Wang JJ, Zhou YB, Han Y, Chen Q, Li YH, et al. NLRP3 inflammasome activation contributes to VSMC phenotypic transformation and proliferation in hypertension. Cell Death Dis. 2017;8:e3074. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Abderrazak A, Syrovets T, Couchie D, El Hadri K, Friguet B, Simmet T and Rouis M. NLRP3 inflammasome: from a danger signal sensor to a regulatory node of oxidative stress and inflammatory diseases. Redox Biol. 2015;4:296–307. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195.Liu Q, Zhang D, Hu D, Zhou X and Zhou Y. The role of mitochondria in NLRP3 inflammasome activation. Mol Immunol. 2018;103:115–124. [DOI] [PubMed] [Google Scholar]
- 196.Shimada K, Crother TR, Karlin J, Dagvadorj J, Chiba N, Chen S, Ramanujan VK, Wolf AJ, Vergnes L, Ojcius DM, et al. Oxidized mitochondrial DNA activates the NLRP3 inflammasome during apoptosis. Immunity. 2012;36:401–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.Sandall CF and MacDonald JA. Effects of phosphorylation on the NLRP3 inflammasome. Arch Biochem Biophys. 2019;670:43–57. [DOI] [PubMed] [Google Scholar]
- 198.Gerdes N, Sukhova GK, Libby P, Reynolds RS, Young JL and Schonbeck U. Expression of interleukin (IL)-18 and functional IL-18 receptor on human vascular endothelial cells, smooth muscle cells, and macrophages: implications for atherogenesis. J Exp Med. 2002;195:245–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199.Nakamura S, Otani T, Okura R, Ijiri Y, Motoda R, Kurimoto M and Orita K. Expression and responsiveness of human interleukin-18 receptor (IL-18R) on hematopoietic cell lines. Leukemia. 2000;14:1052–9. [DOI] [PubMed] [Google Scholar]
- 200.Thomassen E, Bird TA, Renshaw BR, Kennedy MK and Sims JE. Binding of interleukin-18 to the interleukin-1 receptor homologous receptor IL-1Rrp1 leads to activation of signaling pathways similar to those used by interleukin-1. J Interferon Cytokine Res. 1998;18:1077–88. [DOI] [PubMed] [Google Scholar]
- 201.Chandrasekar B, Mummidi S, Mahimainathan L, Patel DN, Bailey SR, Imam SZ, Greene WC and Valente AJ. Interleukin-18-induced human coronary artery smooth muscle cell migration is dependent on NF-kappaB- and AP-1-mediated matrix metalloproteinase-9 expression and is inhibited by atorvastatin. J Biol Chem. 2006;281:15099–109. [DOI] [PubMed] [Google Scholar]
- 202.Dorffel Y, Latsch C, Stuhlmuller B, Schreiber S, Scholze S, Burmester GR and Scholze J. Preactivated peripheral blood monocytes in patients with essential hypertension. Hypertension. 1999;34:113–7. [DOI] [PubMed] [Google Scholar]
- 203.Jimenez-Altayo F, Briones AM, Giraldo J, Planas AM, Salaices M and Vila E. Increased superoxide anion production by interleukin-1beta impairs nitric oxide-mediated relaxation in resistance arteries. J Pharmacol Exp Ther. 2006;316:42–52. [DOI] [PubMed] [Google Scholar]
- 204.Dorrance AM. Interleukin 1-beta (IL-1beta) enhances contractile responses in endothelium-denuded aorta from hypertensive, but not normotensive, rats. Vascul Pharmacol. 2007;47:160–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205.Rabkin SW. The role of interleukin 18 in the pathogenesis of hypertension-induced vascular disease. Nat Clin Pract Cardiovasc Med. 2009;6:192–9. [DOI] [PubMed] [Google Scholar]
- 206.Valente AJ, Yoshida T, Murthy SN, Sakamuri SS, Katsuyama M, Clark RA, Delafontaine P and Chandrasekar B. Angiotensin II enhances AT1-Nox1 binding and stimulates arterial smooth muscle cell migration and proliferation through AT1, Nox1, and interleukin-18. Am J Physiol Heart Circ Physiol. 2012;303:H282–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207.Beltrami-Moreira M, Vromman A, Sukhova GK, Folco EJ and Libby P. Redundancy of IL-1 Isoform Signaling and Its Implications for Arterial Remodeling. PLoS One. 2016;11:e0152474. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208.Omi T, Kumada M, Kamesaki T, Okuda H, Munkhtulga L, Yanagisawa Y, Utsumi N, Gotoh T, Hata A, Soma M, et al. An intronic variable number of tandem repeat polymorphisms of the cold-induced autoinflammatory syndrome 1 (CIAS1) gene modifies gene expression and is associated with essential hypertension. Eur J Hum Genet. 2006;14:1295–305. [DOI] [PubMed] [Google Scholar]
- 209.Bruder-Nascimento T, Ferreira NS, Zanotto CZ, Ramalho F, Pequeno IO, Olivon VC, Neves KB, Alves-Lopes R, Campos E, Silva CA, et al. NLRP3 Inflammasome Mediates Aldosterone-Induced Vascular Damage. Circulation. 2016;134:1866–1880. [DOI] [PubMed] [Google Scholar]
- 210.Dalekos GN, Elisaf M, Bairaktari E, Tsolas O and Siamopoulos KC. Increased serum levels of interleukin-1beta in the systemic circulation of patients with essential hypertension: additional risk factor for atherogenesis in hypertensive patients? J Lab Clin Med. 1997;129:300–8. [DOI] [PubMed] [Google Scholar]
- 211.Ridker PM, Everett BM, Thuren T, MacFadyen JG, Chang WH, Ballantyne C, Fonseca F, Nicolau J, Koenig W, Anker SD, et al. Antiinflammatory Therapy with Canakinumab for Atherosclerotic Disease. N Engl J Med. 2017;377:1119–1131. [DOI] [PubMed] [Google Scholar]
- 212.Ridker PM, MacFadyen JG, Glynn RJ, Koenig W, Libby P, Everett BM, Lefkowitz M, Thuren T and Cornel JH. Inhibition of Interleukin-1beta by Canakinumab and Cardiovascular Outcomes in Patients With Chronic Kidney Disease. J Am Coll Cardiol. 2018;71:2405–2414. [DOI] [PubMed] [Google Scholar]
- 213.Everett BM, Cornel JH, Lainscak M, Anker SD, Abbate A, Thuren T, Libby P, Glynn RJ and Ridker PM. Anti-Inflammatory Therapy With Canakinumab for the Prevention of Hospitalization for Heart Failure. Circulation. 2019;139:1289–1299. [DOI] [PubMed] [Google Scholar]
- 214.Ling YH, Krishnan SM, Chan CT, Diep H, Ferens D, Chin-Dusting J, Kemp-Harper BK, Samuel CS, Hewitson TD, Latz E, et al. Anakinra reduces blood pressure and renal fibrosis in one kidney/DOCA/salt-induced hypertension. Pharmacol Res. 2017;116:77–86. [DOI] [PubMed] [Google Scholar]
- 215.Shirasuna K, Karasawa T, Usui F, Kobayashi M, Komada T, Kimura H, Kawashima A, Ohkuchi A, Taniguchi S and Takahashi M. NLRP3 Deficiency Improves Angiotensin II-Induced Hypertension But Not Fetal Growth Restriction During Pregnancy. Endocrinology. 2015;156:4281–92. [DOI] [PubMed] [Google Scholar]
- 216.Coll RC, Robertson AA, Chae JJ, Higgins SC, Munoz-Planillo R, Inserra MC, Vetter I, Dungan LS, Monks BG, Stutz A, et al. A small-molecule inhibitor of the NLRP3 inflammasome for the treatment of inflammatory diseases. Nat Med. 2015;21:248–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217.Dinh QN, Drummond GR, Kemp-Harper BK, Diep H, De Silva TM, Kim HA, Vinh A, Robertson AAB, Cooper MA, Mansell A, et al. Pressor response to angiotensin II is enhanced in aged mice and associated with inflammation, vasoconstriction and oxidative stress. Aging (Albany NY). 2017;9:1595–1606. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 218.Qi J, Zhao XF, Yu XJ, Yi QY, Shi XL, Tan H, Fan XY, Gao HL, Yue LY, Feng ZP, et al. Targeting Interleukin-1 beta to Suppress Sympathoexcitation in Hypothalamic Paraventricular Nucleus in Dahl Salt-Sensitive Hypertensive Rats. Cardiovasc Toxicol. 2016;16:298–306. [DOI] [PubMed] [Google Scholar]
- 219.Hu Q, Zhang T, Yi L, Zhou X and Mi M. Dihydromyricetin inhibits NLRP3 inflammasome-dependent pyroptosis by activating the Nrf2 signaling pathway in vascular endothelial cells. Biofactors. 2018;44:123–136. [DOI] [PubMed] [Google Scholar]
- 220.Xing JH, Li R, Gao YQ, Wang MY, Liu YZ, Hong J, Dong JZ, Gu HP and Li L. NLRP3 inflammasome mediate palmitate-induced endothelial dysfunction. Life Sci. 2019;239:116882. [DOI] [PubMed] [Google Scholar]
- 221.Chen S, Wang Y, Pan Y, Liu Y, Zheng S, Ding K, Mu K, Yuan Y, Li Z, Song H, et al. Novel Role for Tranilast in Regulating NLRP3 Ubiquitination, Vascular Inflammation, and Atherosclerosis. J Am Heart Assoc. 2020;9:e015513. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 222.Huang W, Huang M, Ouyang H, Peng J and Liang J. Oridonin inhibits vascular inflammation by blocking NF-kappaB and MAPK activation. Eur J Pharmacol. 2018;826:133–139. [DOI] [PubMed] [Google Scholar]
- 223.Zahid A, Li B, Kombe AJK, Jin T and Tao J. Pharmacological Inhibitors of the NLRP3 Inflammasome. Front Immunol. 2019;10:2538. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 224.Ferreira NS, Bruder-Nascimento T, Pereira CA, Zanotto CZ, Prado DS, Silva JF, Rassi DM, Foss-Freitas MC, Alves-Filho JC, Carlos D, et al. NLRP3 Inflammasome and Mineralocorticoid Receptors Are Associated with Vascular Dysfunction in Type 2 Diabetes Mellitus. Cells 2019;8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 225.Pereira CA, Carlos D, Ferreira NS, Silva JF, Zanotto CZ, Zamboni DS, Garcia VD, Ventura DF, Silva JS and Tostes RC. Mitochondrial DNA Promotes NLRP3 Inflammasome Activation and Contributes to Endothelial Dysfunction and Inflammation in Type 1 Diabetes. Front Physiol. 2019;10:1557. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 226.Ferrucci L and Fabbri E. Inflammageing: chronic inflammation in ageing, cardiovascular disease, and frailty. Nat Rev Cardiol. 2018;15:505–522. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 227.Yin Y, Zhou Z, Liu W, Chang Q, Sun G and Dai Y. Vascular endothelial cells senescence is associated with NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome activation via reactive oxygen species (ROS)/thioredoxin-interacting protein (TXNIP) pathway. Int J Biochem Cell Biol. 2017;84:22–34. [DOI] [PubMed] [Google Scholar]
- 228.Marin-Aguilar F, Lechuga-Vieco AV, Alcocer-Gomez E, Castejon-Vega B, Lucas J, Garrido C, Peralta-Garcia A, Perez-Pulido AJ, Varela-Lopez A, Quiles JL, et al. NLRP3 inflammasome suppression improves longevity and prevents cardiac aging in male mice. Aging Cell. 2020;19:e13050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 229.Wu X, Hakimi M, Wortmann M, Zhang J, Bockler D and Dihlmann S. Gene expression of inflammasome components in peripheral blood mononuclear cells (PBMC) of vascular patients increases with age. Immun Ageing. 2015;12:15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 230.Bi X, Zhang G, Wang X, Nguyen C, May Herman I, Li X, Al-Hashimi Ali A, Austin Richard C, Gillette Thomas G, Fu G, et al. Endoplasmic Reticulum Chaperone GRP78 Protects Heart From Ischemia/Reperfusion Injury Through Akt Activation. Circulation Research. 2018;122:1545–1554. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 231.Blackwood EA, Azizi K, Thuerauf DJ, Paxman RJ, Plate L, Kelly JW, Wiseman RL and Glembotski CC. Pharmacologic ATF6 activation confers global protection in widespread disease models by reprograming cellular proteostasis. Nat Commun. 2019;10:187. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 232.Rothman AM, MacFadyen J, Thuren T, Webb A, Harrison DG, Guzik TJ, Libby P, Glynn RJ and Ridker PM. Effects of Interleukin-1beta Inhibition on Blood Pressure, Incident Hypertension, and Residual Inflammatory Risk: A Secondary Analysis of CANTOS. Hypertension. 2020;75:477–482. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 233.Tong Y, Wang Z, Cai L, Lin L, Liu J and Cheng J. NLRP3 Inflammasome and Its Central Role in the Cardiovascular Diseases. Oxid Med Cell Longev. 2020;2020:4293206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 234.Kawaguchi M, Takahashi M, Hata T, Kashima Y, Usui F, Morimoto H, Izawa A, Takahashi Y, Masumoto J, Koyama J, et al. Inflammasome activation of cardiac fibroblasts is essential for myocardial ischemia/reperfusion injury. Circulation. 2011;123:594–604. [DOI] [PubMed] [Google Scholar]
