Open Access

The role of heat shock proteins in the pathogenesis of heart failure (Review)

  • Authors:
    • Anastasia Pavlovna Sklifasovskaya
    • Mikhail Blagonravov
    • Anna Ryabinina
    • Vyacheslav Goryachev
    • Sergey Syatkin
    • Sergey Chibisov
    • Karina Akhmetova
    • Daniil Prokofiev
    • Enzo Agostinelli
  • View Affiliations

  • Published online on: September 27, 2023     https://doi.org/10.3892/ijmm.2023.5309
  • Article Number: 106
  • Copyright: © Sklifasovskaya et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The influence of heat shock proteins (HSPs) on protein quality control systems in cardiomyocytes is currently under investigation. The effect of HSPs on the regulated cell death of cardiomyocytes (CMCs) is of great importance, since they play a major role in the implementation of compensatory and adaptive mechanisms in the event of cardiac damage. HSPs mediate a number of mechanisms that activate the apoptotic cascade, playing both pro‑ and anti‑apoptotic roles depending on their location in the cell. Another type of cell death, autophagy, can in some cases lead to cell death, while in other situations it acts as a cell survival mechanism. The present review considered the characteristics of the expression of HSPs of different molecular weights in CMCs in myocardial damage caused by heart failure, as well as their role in the realization of certain types of regulated cell death.

1. Introduction

The reliability of cellular protein composition is important for proper cellular function in various tissues, especially in the myocardium, as cardiac myocytes (CMCs) are terminally differentiated cells with very limited regenerative potential (1). At the same time, the metabolic demands of the heart require a tight control of protein quality (2). Cellular stress in the myocardium, which occurs during ischemia (3), hypertension (4) and metabolic disorders including diabetes mellitus (DM) (5), can disrupt protein homeostasis and cause abnormal folding of cellular proteins. Intracellular accumulation of toxic, misfolded proteins and their aggregates may contribute to the development of heart failure (6). Cells have an innate mechanism for detecting protein misfolding, which they can use to repair or remove these proteins. This mechanism is called 'protein quality control' and involves three main pathways (Fig. 1) (7,8). First, misfolded proteins are exposed to heat shock proteins (HSPs), which are characterized as molecular chaperones. If the chaperone system fails to refold denatured proteins, they are ubiquitinated by an activated second system and delivered to the proteasome mechanism for subsequent degradation. When these systems are disrupted or overloaded, such ubiquitinated proteins accumulate in agresomes, perinuclear structures which are ultimately utilized by autophagy, the third mechanism (9-12).

HSPs are a family of conserved proteins that can contribute to proper protein folding, maintain protein stability and act as molecular chaperones to regulate cellular metabolism, tissue composition and other processes (13). HSPs are also called stress-induced proteins because of their ability to stabilize and repair proteins during their synthesis in the cell under the action of unfavorable agents on the cell (14,15). Meanwhile, they are also involved in the modulation of inflammatory response, oxidative stress and metabolism (16-18).

The effect of HSPs on the regulated cell death of CMCs is of great interest, as they contribute significantly to the implementation of compensatory and adaptive mechanisms, such as in the case of cardiac damage (19-21). HSPs mediate a number of activation mechanisms of the apoptotic cascade, playing both pro- and anti-apoptotic roles depending on their cellular localization (22). In some cases, autophagy can lead to cell death, while in other cases it acts as a mechanism for their survival (23,24).

Despite the availability of numerous data, concerning the role of HSPs and factors involved in programmed cell death in response to pathological agents, there is currently no clear idea of their role in the pathogenesis of heart failure caused by various types of cardiovascular pathology.

2. The role of HSPs in the structural and metabolic changes of CMCs in various types of cardiovascular pathology

HSPs and inflammation

HSPs are involved in inflammation, which mediates various pathological processes in the myocardium. Schroder and Tschopp (16) and other scientists paid special attention to the role of HSPs in the formation of intracellular platforms; inflammasomes, which are activated by 'danger' signals and induce inflammatory caspases, mainly caspase-1, which contributes to the production of pro-inflammatory cytokine IL-1β (25,26). A number of other cytokines from the family of pro-inflammatory mediators are also related to these processes (27). TNF is associated with the induction of necrosis, cell damage and tearing of membrane molecules, membrane modularity (28,29). Some members of the HSP family support inflammasome activation, while others inhibit it. In particular, HSP60 (Fig. 2) is required for the phosphorylation and nuclear localization of NF-kB after stimulation by IL-1β in microglia (30). Knockdown of the HSP60 gene leads to inhibition of phosphorylation of the p65 NF-kB subunit and consequently to suppression of NF-kB nuclear translocation. Presumably, HSP60 promotes p65 phosphorylation (31). This activation of the NF-kB pathway leads to the overexpression of both pro-IL-1β and nucleotide-binding oligomerization domain-containing protein 3 (NLRP3), corresponding to the 'priming' step of NLRP3 inflammasome activation (32). HSP60 also induces mitochondrial damage, as evidenced by a decrease in their membrane potential after HSP60 overexpression and treatment with IL-1β. This is accompanied by an increase in reactive oxygen species (ROS) production, which contributes to oxidative stress, which in turn activates the NLRP3 inflammasome (33).

HSPs and platelets, thrombosis

Platelets serve as regulators of hemostasis. Upon interaction with extracellular matrix proteins, such as collagen, platelets are activated, leading to shape change, secretion, filopodia formation and ultimately aggregation (34-36). Platelet aggregation is largely mediated by activation of the platelet integrin αIIbβ3, which undergoes conformational changes in response to stimulation of soluble fibrinogen binding (37,38). The role of HSPs in the activation of thrombus formation is of great interest, particularly in relation to the problem of preventing coronary artery thrombosis.

Rigg et al (39) demonstrated the role of HSP70 in the modulation of integrin activation. The regulation of integrin and platelet conformational changes by molecular chaperones together with thiol isomerases has been suggested. The study demonstrated the importance of intracellular HSP70 in the regulation of integrin activation and platelet secretion, but the involvement of extracellular HSP70 in the modulation of platelet integrin activity and thrombus formation should also be considered (39). Activated HSP70 can be translocated to the cell surface and secreted by exosomes. In the extracellular space, HSP70 is able to bind Toll-like and other receptors on monocytes, macrophages, dendritic cells, followed by activation of NFκB/MAP kinase pathways, initiating an inflammatory response in tissues (40-42).

HSP and arterial hypertension (including in combination with DM)

In a recent study by Blagonravov et al (43), it was found that the production of HSPs can either increase or decrease under the action of various stress factors. In particular, the expression of HSP60 in CMCs of left ventricular (LV) myocardium decreased both during hemodynamic overload of the LV caused by hypertension and during metabolic disturbances in the myocardium associated with DM. Moreover, the combination of hypertension and DM was not associated with a synergistic negative effect on this process. On the contrary, a slightly less pronounced inhibition of HSP60 production was observed. Suppression of HSP60 synthesis is most likely to be associated with energy deprivation due to LV overload and/or insulin-dependent DM, since this protein is ATP-dependent.

HSPs and metabolic syndrome

Metabolic syndrome is a risk factor for the development of heart failure and DM. The features of the metabolic syndrome are systemic inflammation and oxidative stress, which can enhance the expression and release of HSP (44). HSPs play a role in cell signaling and regulation of cell metabolism in conditions of insulin resistance (45). In obesity, an uncontrolled inflammatory reaction and a disorder of the body's defense system play an important role in inhibiting the signaling cascade of insulin receptors and, as a consequence, a disorder of systemic metabolic homeostasis (41). Regulation of HSP expression at the gene level is an important aspect of HSP72 activity in metabolic syndrome. Overexpression of HSP72 leads to an increase in the number of mitochondria in cells, the oxidative capacity and the sensitivity of cells to insulin (44,45). Similarly, lack of HSP72 expression results in mitochondrial dysfunction and insulin resistance (46,47). In addition to increasing HSP72 levels and consequently the ability to improve mitochondrial quality control, exercise also contributes to an increase in the expression of peroxisome proliferator-activated receptor γ-coactivator 1-α (PGC1a) (48). PGC1a is the major transcriptional coactivator for mitochondrial formation (49). The upstream regulatory elements of the PPARGC1A gene were found to contain the heat shock element (HSE) binding sequence (50). This HSE sequence provides a docking site for the primary HSP transcription factor, heat shock factor 1 (HSF1) (51). Numerous HSF1 gene activation and knockdown experiments have convincingly demonstrated that HSF1 is the master regulator of mitochondrial biogenesis, enzymatic function and whole-body metabolism (48). These data illustrate a coordination of HSF1 downstream targets (HSP1 and PGC1a) in the regulation of mitochondrial biogenesis, quality control and enzymatic function in metabolic demand and/or chronic disease (52).

HSPs and myocardial ischemia

Elevated blood levels of HSPs have been found in patients with coronary artery disease (CAD), although the source of these proteins is still controversial (53). In general, the expression of HSPs appears to be increased in response to ischemia and their dysregulated production contributes to the development of cardiovascular disease (54-57).

Extracellular HSP70 has been found to be an independent predictive marker of mortality in patients with progressive heart failure or sudden cardiac death (58-60). Its role in the development of hypertension-induced cardiac hypertrophy and myocardial fibrosis has also been demonstrated (61-63). It is also notable that HSP70 is a ligand for damage-associated molecular pattern receptors, which can induce inflammation in the myocardium (64,65).

The properties of extracellular HSP90 have been demonstrated in a study by Ranek et al (6) in the context of progressive LV hypertrophy. In pathological cardiac hypertrophy, LV mass increases in parallel with extracellular matrix deposition, followed by fibrosis and heart failure (66-68). The main mediator of this process is TGF-β, which is secreted by CMCs and acts on collagen-secreting fibroblasts (69-71). Extracellular HSP90 appears to play some role in stabilizing the signal of TGF-β by influencing the cascade of processes mediating TGF-β induction (72-74). Inhibition of extracellular HSP90 reduces collagen production and stimulation of the canonical TGF-ß pathway (54,74). Since fibrosis is one of the major factors influencing the pathogenesis of a number of chronic cardiac diseases, including CAD and heart failure, targeting extracellular HSP90 can be considered an important point of application for therapeutic intervention with functions different from those characteristics of the intracellular pool of this particular HSP.

3. Features of the induction of small ATP-independent HSPs in myocardial damage

Small HSPs are ATP-independent proteins and represent the first line of defense in preventing intracellular protein aggregation under cellular stress when denaturation is highly activated (75,76). At the same time, high molecular weight HSPs, such as HSP60, HSP70 and HSP90, bind unfolded or misfolded proteins and promote their refolding using the energy of ATP hydrolysis (77-79). Therefore, under cellular stress and energy deprivation, ATP-independent small HSPs are able to rapidly and metabolically prevent protein aggregation, maintain denaturing proteins in a folded state and inhibit the denaturation process until ATP-dependent HSPs complete the refolding process (80-82). If proper folding does not occur, small HSPs contribute to the clearance of denatured proteins by directing them to the pathway of degradation mechanisms (Fig. 3) (83-85).

Currently, the role of HSPs in inflammation and their antioxidant capacity are of considerable interest, especially in the aspect of modulating the progression of atherosclerosis by altering its inflammatory mechanisms (86-88).

HSPs are also activated in response to the appearance of oxidized low-density lipoproteins and mediate an anti-inflammatory mechanism through the release of pro-inflammatory IL-10 as well as through the activation of NFκB (89-91). The intracellular function of the HSP27 chaperone is regulated by phosphorylation and dephosphorylation in large aggregates that modulate the assembly of an ATP-independent network. As a chaperone, HSP27 is involved in DNA stabilization, supports antioxidant responses and also acts as an anti-apoptotic factor (92-94). Extracellular release of HSP27 from tissues where blood vessels are affected by atherosclerosis may result from cellular injury or occur in association with secretory lysosomes or exosomes. In its extracellular location, HSP27 binds to a variety of cell membrane receptors on endothelial and immunocompetent cells, including CD91, CD40, CD36, CD14, scavenger receptor A (SR-A) and Toll-like receptors (TLRs) (95,96). The recombinant receptor HSP27 induces TLR-mediated NFκB activation with secretion of both pro- and anti-inflammatory cytokines (55). According to the available data, HSP27 provides protection against the progression of atherosclerosis (22,97).

In a recently published study by Sklifasovskaya and Blagonravov (98), the results of assessment of HSP10 and HSP27 expression in rat LV myocardium in hypertension, DM and their combination were presented. In case of combination of hemodynamic overload and DM, small HSPs showed different expression levels in CMCs. This may be due to activation of different pathways of cell protection and damage. In particular, HSP27 expression increased only in arterial hypertension (AH) of longer duration, while the increase in HSP10 expression was observed in combined pathology. Thus, the levels of ATP-independent HSP10 and HSP27 increase in certain types of myocardial alterations associated with energy deficit. Expression of HSP10 and HSP27 in CMCs changed inversely in AH of different duration, insulin-dependent DM and their combination. Protein HSP27 may play a more important role in cardioprotection in long-term hypertension and protein HSP10 in the case of the combination of AH and DM (98).

Induction of mitochondrial (mt) ROS during hyperglycemia is a key event responsible for endothelial activation and damage (99). HSP22 has been shown to protect vascular endothelium from hyperglycemia-induced damage by reducing mtROS production (100-103). Yu et al (101) performed a series of studies using a high fat diet and a streptozotocin model to induce DM. They also exposed human umbilical vein endothelial cells to a high concentration of glucose after overexpressing or silencing HSP22 to investigate the role of the latter. It was found that HSP22 significantly reduced endothelial cell activation and vascular lesions by inhibiting endothelial adhesion, suppressing mtROS-mediated endothelial activation and injury, abolishing a hyperglycemia-induced increase in mtROS and also reducing cytokine secretion. In addition, HSP22 attenuated mtROS and mitochondrial dysfunction in hyperglycemia-stimulated endothelial cells (101).

4. The role of HSPs in regulated cell death

The role of HSPs in autophagy

An important role in the process of autophagy is its regulation by HSPs (104-106). During heat shock, HSP production and autophagy activation are reactions aimed at maintaining the quality of cellular proteins and they complement each other under cellular stress (107,108). Heat stress can lead to protein aggregation and contribute to the development of dysfunction of individual elements of the intracellular environment (10,109-111). Insufficient autophagy or partial degradation of damaged organelles leads to the generation of ROS and the loss of the major functions of lysosomes, resulting in cell death (112-114). In addition, inhibition of autophagy has been observed during the development of myocardial hypertrophy in response to deletion of the Atg5 gene in mouse CMCs (115).

In the case of cardiovascular pathology, cellular stress may also be due to cardiac overload associated with increased resistance to cardiac output caused by hypertension or aortic stenosis. An increase in the autophagic flux of CMCs occurs in association with their hypertrophy (116,117). Myocardial ischemia is associated with a transient increase in autophagic flux, which, however, decreases over time and falls below normal baseline levels (19,118-120). In a model of acute focal LV ischemia, it was shown that the content of Beclin-1 in the CMC cytoplasm was significantly increased on day 1 and later, on days 3 and 5, it gradually decreased but remained above the control level (20). In ischemia, the intensification of CMC autophagy is aimed at replenishing metabolic substrates and removing damaged organelles (121-123). Nutrient depletion stimulates AMP-activated protein kinase, which in turn inhibits mTOR, thereby removing the main inhibitory factor in this process (124-126). In this context, autophagy is considered to be an adaptive response to ischemic injury with cardioprotective effects. Thus, targeted activation of autophagy may be a potential therapeutic approach to heat stress-induced cardiovascular dysfunction (23).

A study showed that HSPB6 regulates the ubiquitination and proteasomal degradation of BECN1. This effect appears to be mediated by a direct interaction of HSPB6 with BECN1 (127). These ideas concerning the role of HSPB6 in autophagy were generated in the context of the description of a novel human mutation in the gene encoding HSPB6 (HSPB6S10F) identified in patients with dilated cardiomyopathy (DCM) (128,129). Negative effects of HSPB6S10F were associated with autophagy dysregulation. It is known that constitutive autophagy in the heart under basal conditions is a homeostatic mechanism aimed at maintaining myocyte size, structure and function of the heart (130). It has also been found that the activity of autophagy was significantly reduced in CMCs with the mutation of the HSPB6S10F gene, as evidenced by the decreased number of autophagosomes and inhibition of autophagic flux. Under these conditions, the rate of CMC apoptosis increases and hypertrophic remodeling develops, ultimately contributing to the progression of heart failure (127). Similar negative effects were associated with a decrease in the interaction of mutant HSPB6S10F with Beclin1 (BECN1), leading to BECN1 ubiquitation and its degradation by the proteasome. As a result, autophagy flux is significantly suppressed and CMC apoptosis is enhanced. Conversely, overexpression of wild-type HSPB6 (HSPB6 WT) contributed to increased BECN1 levels and also competitively repressed BECN1 binding to Bcl2, thereby stimulating autophagy (127). These data reveal a novel regulatory mechanism by which HSPB6 promotes cell survival through its interaction with BECN1.

Depending on the specific cellular conditions, autophagy can both protect cells from death and act as a means of cellular self-destruction (131,132). The protective mechanism of autophagy is implemented as follows: Organelles and/or parts of the cytoplasm are engulfed by double-membrane autophagic vacuoles, resulting in the physiological utilization of old or damaged organelles (133,134). This provides cells with metabolic substrates to meet energy demands during cellular stress (135,136). However, the accumulation of these vacuoles and further activation of the autophagic pathway represents an alternative mechanism of cell death by atrophy and functional collapse (type II cell death) (137-139). Autophagy can also activate apoptotic (type I) or necrotic (type III) cell death programs by activating common regulators such as Bcl-2 family proteins (140,141).

In a recently published study by Blagonravov et al (142), the role of BECN1-dependent autophagy was evaluated in hypertension, DM and their combination. The activation of Bax and Bcl-2 was also examined. It was shown that chronic hypertensive overload of the LV is associated with a decrease in the rate of CMC autophagy. In isolated DM and its combination with hypertension, the opposite effect was observed, which was manifested as a significant increase in the basal level of BECN1-dependent autophagy. In the light of the concept of protein quality control, this phenomenon can be considered as a mechanism for the survival of damaged cells. In hypertension and isolated DM, there is an induction of the apoptotic cascade in LV CMCs, as indicated by an increase in Bax expression and a decrease in the Bcl-2/Bax ratio. At the same time, in the combined pathology, the level of Bax does not increase as significantly as in the other groups, but it remains higher than in the control group. The level of Bcl-2 was also significantly increased and the Bcl-2/Bax ratio tended to increase, but remained below the control. This indicates a decrease of the apoptotic cascade in the comorbid pathology compared to isolated hypertension and DM (142,143).

According to the results obtained by Blagonravov et al (43), it is possible to reveal the role of HSP60 and its co-chaperone expression in DM. In particular, the level of BECN1 was increased in the DM group relative to HSP60/HSP10 and in the combined hypertension and DM group, the HSP60/HSP10 chaperone complex and BECN1 protein were expressed at the same level.

This fact indicates that the activation of these proteins plays a maladaptive role in the activity of CMCs, which can serve as an important marker for assessing the progression of vascular damage in this type of pathology. A decrease in the expression of the HSP60/HSP10 complex relative to Bax was also found in all the pathological models studied, while this difference was less pronounced in the combined hypertension with DM group (43,98,143).

Thus, a decrease in the production of HSP60 can be considered as one of the pathophysiological mechanisms of LV myocardial damage caused by hypertension and/or DM. In the mentioned study, the expression of HSP27 in LV myocardium was also evaluated. In the group with longer duration of hypertension (SHR rats aged 57 weeks), the ratio of Bax and Bcl-2 was the most pronounced, while the level of HSP27 was also the highest in all groups, which may indicate the activation of protein defense mechanisms directed to Bax binding (43,98,143).

It can be concluded that the role of HSP in autophagy regulation and modulation of the apoptotic cascade requires further investigation.

The role of HSP in apoptosis

Apoptosis, or programmed cell death, is characterized by the activation of the caspase cascade: 'Initiator' caspases induce a chain reaction of specific 'effector' caspases (144-146). These, in turn, are cleaved and thus activate each other. There are two main pathways of initiator caspase activation (147,148). The extrinsic pathway is associated with 'death receptors' on the cell surface, while the intrinsic pathway is induced by pro-apoptotic factors such as cytochrome c released from mitochondria. Cytochrome c, which binds to apoptotic protease activating factor 1 (Apaf-1) and pro-caspase-9, forms an apoptosome that stimulates the activation of caspase-9, which then activates the 'effector' caspase-3 and initiates the apoptotic protease cascade (149,150).

HSPs have a wide range of functions in apoptotic processes. Most of them are aimed at their suppression (151,152). Notably, the same cellular stress signals that induce apoptosis also stimulate HSP synthesis and release. However, when HSP production is increased, apoptosis is inhibited due to the suppression of pro-apoptotic factors such as р53, Bax, Bid, Akt, Apaf-1 and other members of the Bcl-2 family (Fig. 4). HSPs promote cell survival by protecting cells from changes in cellular redox homeostasis and stabilizing the cytoskeleton (152). In addition, HSPs can directly inhibit several steps of the apoptotic pathway (153). HSPs also inhibit the release of pro-apoptotic molecules from mitochondria, thereby reducing caspase activation (151,154-156).

During the execution of the apoptotic cascade and under ATP deprivation, HSP60 protects mitochondrial proteins, facilitates their folding and prevents their degradation (157,158). In particular, when the Hsp60 gene is deleted in CMCs of young mice, HSP60-dependent mitochondrial proteins are degraded by LONP1 and mitochondrial dysfunction develops, which is one of the mechanisms of DCM and heart failure (159). Cytosolic HSP60 co-localizes with Bax and plays an anti-apoptotic role in cardiac myocytes. Loss of cytosolic HSP60 induces mitochondrial Bax translocation, cytochrome c release and caspase-3 activation, leading to apoptotic cell death (30). In addition, hypoxia triggers apoptosis by inducing dissociation of the HSP60-Bax complex through translocation of cytosolic HSP60 to the membrane and Bax to the mitochondria (160).

It is also worth considering the possibility of increasing the level of Bcl-2 in cells and reducing the activation of the apoptotic cascade by HSPB1. Tian et al (161) showed that HSP27 can attenuate oxidative stress-induced apoptosis in endothelial cells by increasing the level of Bcl-2 and decreasing the content of cleaved caspase-3 and Bax. In addition, under ER stress, HSP27 promotes ERK-mediated phosphorylation and Bim degradation by inhibiting the mechanisms of the intrinsic pathway of apoptosis initiation (162). HSP27 can also directly bind to cytochrome c in the cytosol, inhibiting apoptosome formation and interfering with downstream caspase activation. HSP27 can also suppress apoptosis by inhibiting mitochondrial Smac release and subsequent activation of the descending caspase cascade (161). The anti-apoptotic properties of HSP27 are attributed to its direct interaction with caspase-3. HSP27 binds to the pro-domain of caspase-3 and inhibits its proteolytic activation (161,163).

The role of HSP70 in the initiation of the apoptotic cascade has also been demonstrated in a model of myocardial ischemia/reperfusion injury (164-166). Overexpression of HSP70 in the myocardium and endothelium has a cardio-protective effect and increases myocardial injury tolerance. These mechanisms are associated with inhibition of apoptosis and oxidative stress and improvement of endothelial function (60,167). Myocardial HSP70 activates mitochondrial superoxide dismutase Mn-SOD and inhibits nuclear translocation of phosphorylated eukaryotic elongation factor 2 and apoptosis inducing factor, resulting in improvement of mitochondrial function and suppression of apoptosis (168,169). In addition, an increase in mitochondrial aldehyde dehydrogenase 2 activity triggers the accumulation of 4-hydroxynonenal during ischemic myocardial injury, which initiates pro-apoptotic signaling by reducing HSP70 and activating the JNK/p53 pathway. This mechanism ultimately contributes to the development of heart failure, whereas the aforementioned process can be reversed by overexpression of HSP70 (170). It has also been shown that intracellular HSP70 has a cardioprotective effect, whereas extracellular HSP70 appears to be pro-apoptotic (171).

Thus, cytosolic HSPs protect the myocardium from ischemia/reperfusion injury by inhibiting the activation of the apoptotic cascade in CMCs. In particular, the intracellular or extracellular location of HSPs is very important for their function in myocardial infarction or ischemia-reperfusion. Intracellular HSPs have cardio-protective properties, whereas extracellular HSPs exhibit cardio-toxic effects during ischemia-reperfusion.

5. Conclusions and perspectives

HSPs are an important component of the protein quality control system in the cell, both under normal and pathological conditions. They ensure the correct assembly of a number of intracellular proteins and regulate a significant part of the synthesis processes. Insufficient or excessive production of HSPs leads to disruption of cell homeostasis and may also contribute to the activation of cellular stress. This can result in endoplasmic reticulum damage, mitochondrial dysregulation and modulation of regulated (programmed) cell death, including apoptosis and autophagic flux.

Numerous studies (6,21,41,43,44,56-64) confirm a significant increase or decrease in myocardial HSP production in a variety of cardiac diseases, heat stress and metabolic disorders. Recently, the possibility of inducing HSPs to protect myocardial cells during oxidative stress caused by energy deprivation has been widely discussed. This approach is considered as a therapeutic method in the treatment of cardiovascular diseases of various origins, including DM.

Understanding the mechanisms of action of HSPs in the cardiovascular system can serve as a basis for the development of new methods of pharmacotherapy of cardiac pathology.

Availability of data and materials

Data sharing is not applicable to this article, as no data sets were generated or analyzed during the current study.

Authors' contributions

AS and MB conceived the study and wrote the original draft. SS, SC, AR, KA, DP, VG and EA participated in writing and editing the manuscript. SS and EA edited the manuscript. All the authors read and approved the final manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgments

The authors thank the International Polyamines Foundation ETS-ONLUS for their helpful suggestions and assistance in reading the revised manuscripts.

Funding

The present study was supported by project no. 032532-0-000 of RUDN University, Moscow, Russia.

References

1 

Safari S, Malekvandfard F, Babashah S, Alizadehasl A, Sadeghizadeh M and Motavaf M: Mesenchymal stem cell-derived exosomes: A novel potential therapeutic avenue for cardiac regeneration. Cell Mol Biol (Noisy-le-grand). 62:66–73. 2016.PubMed/NCBI

2 

Tarone G and Brancaccio M: Keep your heart in shape: Molecular chaperone networks for treating heart disease. Cardiovasc Res. 102:346–361. 2014. View Article : Google Scholar : PubMed/NCBI

3 

Rabinovich-Nikitin I, Rasouli M, Reitz CJ, Posen I, Margulets V, Dhingra R, Khatua TN, Thliveris JA, Martino TA and Kirshenbaum LA: Mitochondrial autophagy and cell survival is regulated by the circadian Clock gene in cardiac myocytes during ischemic stress. Autophagy. 17:3794–3812. 2021. View Article : Google Scholar : PubMed/NCBI

4 

Cicalese SM, da Silva JF, Priviero F, Webb RC, Eguchi S and Tostes RC: Vascular stress signaling in hypertension. Circ Res. 128:969–992. 2021. View Article : Google Scholar : PubMed/NCBI

5 

Ma T, Huang X, Zheng H, Huang G, Li W, Liu X, Liang J, Cao Y, Hu Y and Huang Y: SFRP2 improves mitochondrial dynamics and mitochondrial biogenesis, oxidative stress, and apoptosis in diabetic cardiomyopathy. Oxid Med Cell Longev. 2021:92650162021. View Article : Google Scholar : PubMed/NCBI

6 

Ranek MJ, Stachowski MJ, Kirk JA and Willis MS: The role of heat shock proteins and co-chaperones in heart failure. Philos Trans R Soc Lond B Biol Sci. 373:201605302018. View Article : Google Scholar

7 

Maejima Y: The critical roles of protein quality control systems in the pathogenesis of heart failure. J Cardiol. 75:219–227. 2020. View Article : Google Scholar

8 

Schwabl S and Teis D: Protein quality control at the Golgi. Curr Opin Cell Biol. 75:1020742022. View Article : Google Scholar : PubMed/NCBI

9 

Wang X and Robbins J: Heart failure and protein quality control. Circ Res. 99:1315–1328. 2006. View Article : Google Scholar : PubMed/NCBI

10 

Brownstein AJ, Ganesan S, Summers CM, Pearce S, Hale BJ, Ross JW, Gabler N, Seibert JT, Rhoads RP, Baumgard LH and Selsby JT: Heat stress causes dysfunctional autophagy in oxidative skeletal muscle. Physiol Rep. 5:e133172017. View Article : Google Scholar : PubMed/NCBI

11 

Hagymasi AT, Dempsey JP and Srivastava PK: Heat-shock proteins. Curr Protoc. 2:e5922022. View Article : Google Scholar : PubMed/NCBI

12 

Tedesco B, Vendredy L, Timmerman V and Poletti A: The chaperone-assisted selective autophagy complex dynamics and dysfunctions. Autophagy. 19:1619–1641. 2023. View Article : Google Scholar : PubMed/NCBI

13 

Yun CW, Kim HJ, Lim JH and Lee SH: Heat Shock Proteins: Agents of cancer development and therapeutic targets in anti-cancer therapy. Cells. 9:602019. View Article : Google Scholar : PubMed/NCBI

14 

Haslbeck M and Vierling E: A first line of stress defense: Small heat shock proteins and their function in protein homeostasis. J Mol Biol. 427:1537–1548. 2015. View Article : Google Scholar : PubMed/NCBI

15 

Jacob P, Hirt H and Bendahmane A: The heat-shock protein/chaperone network and multiple stress resistance. Plant. Biotechnol J. 15:405–414. 2017. View Article : Google Scholar :

16 

Schroder K and Tschopp J: The inflammasomes. Cell. 140:821–832. 2010. View Article : Google Scholar : PubMed/NCBI

17 

Gomez-Pastor R, Burchfiel ET, Neef DW, Jaeger AM, Cabiscol E, McKinstry SU, Doss A, Aballay A, Lo DC, Akimov SS, et al: Abnormal degradation of the neuronal stress-protective transcription factor HSF1 in Huntington's disease. Nat Commun. 8:144052017. View Article : Google Scholar : PubMed/NCBI

18 

Dowell J, Elser BA, Schroeder RE and Stevens HE: Cellular stress mechanisms of prenatal maternal stress: Heat shock factors and oxidative stress. Neurosci Lett. 709:1343682019. View Article : Google Scholar : PubMed/NCBI

19 

Xie M, Kong Y, Tan W, May H, Battiprolu PK, Pedrozo Z, Wang ZV, Morales C, Luo X, Cho G, et al: Histone deacetylase inhibition blunts ischemia/reperfusion injury by inducing cardiomyocyte autophagy. Circulation. 129:1139–1151. 2014. View Article : Google Scholar : PubMed/NCBI

20 

Blagonravov ML, Korshunova AY, Azova MM, Bondar' SA and Frolov VA: Cardiomyocyte autophagia and morphological alterations in the left ventricular myocardium during acute focal ischemia. Bull Exp Biol Med. 160:398–400. 2016. View Article : Google Scholar : PubMed/NCBI

21 

Zhang HL, Jia KY, Sun D and Yang M: Protective effect of HSP27 in atherosclerosis and coronary heart disease by inhibiting reactive oxygen species. J Cell Biochem. 120:2859–2868. 2019. View Article : Google Scholar

22 

Shan R, Liu N, Yan Y and Liu B: Apoptosis, autophagy and atherosclerosis: Relationships and the role of Hsp27. Pharmacol Res. 166:1051692021. View Article : Google Scholar

23 

Kovaleva OV, Shitova MS and Zborovskaya IB: Autophagy: Cell death or a way of survival? Clin Oncohematology. 7:103–113. 2014.

24 

Del Re DP, Amgalan D, Linkermann A, Liu Q and Kitsis RN: Fundamental mechanisms of regulated cell death and implications for heart disease. Physiol Rev. 99:1765–1817. 2019. View Article : Google Scholar : PubMed/NCBI

25 

Martine P and Rébé C: Heat shock proteins and inflammasomes. Int J Mol Sci. 20:45082019. View Article : Google Scholar : PubMed/NCBI

26 

Choudhury A, Bullock D, Lim A, Argemi J, Orning P, Lien E, Bataller R and Mandrekar P: Inhibition of HSP90 and activation of HSF1 diminish macrophage NLRP3 inflammasome activity in alcohol-associated liver injury. Alcohol Clin Exp Res. 44:1300–1311. 2020. View Article : Google Scholar : PubMed/NCBI

27 

Jurisic V: Multiomic analysis of cytokines in immuno-oncology. Expert Rev Proteomics. 17:663–674. 2020. View Article : Google Scholar : PubMed/NCBI

28 

Jurisic V, Srdic-Rajic V, Konjevic G, Bogdanovic G and Colic M: TNF-α induced apoptosis is accompanied with rapid CD30 and slower CD45 shedding from K-562 cells. J Membr Biol. 239:115–122. 2011. View Article : Google Scholar : PubMed/NCBI

29 

Jurisic V, Terzic T, Colic S and Jurisic M: The concentration of TNF-alpha correlate with number of inflammatory cells and degree of vascularization in radicular cysts. Oral Dis. 14:600–605. 2008. View Article : Google Scholar : PubMed/NCBI

30 

Swaroop S, Sengupta N, Suryawanshi AR, Adlakha YK and Basu A: HSP60 plays a regulatory role in IL-1β-induced microglial inflammation via TLR4-p38 MAPK axis. J Neuroinflammation. 13:272016. View Article : Google Scholar

31 

Li XL, Wang YL, Zheng J, Zhang Y and Zhang XF: Inhibiting expression of HSP60 and TLR4 attenuates paraquat-induced microglial inflammation. Chem Biol Interact. 299:179–185. 2019. View Article : Google Scholar

32 

Kelley N, Jeltema D, Duan Y and He Y: The NLRP3 Inflammasome: An overview of mechanisms of activation and regulation. Int J Mol Sci. 20:33282019. View Article : Google Scholar : PubMed/NCBI

33 

Swaroop S, Mahadevan A, Shankar SK, Adlakha YK and Basu A: HSP60 critically regulates endogenous IL-1β production in activated microglia by stimulating NLRP3 inflammasome pathway. J Neuroinflammation. 15:1772018. View Article : Google Scholar

34 

Aslan JE and McCarty OJ: Rho GTPases in platelet function. J Thromb Haemost. 11:35–46. 2013. View Article : Google Scholar

35 

Elvers M: RhoGAPs and Rho GTPases in platelets. Hamostaseologie. 36:168–177. 2016. View Article : Google Scholar

36 

Ngo ATP, Parra-Izquierdo I, Aslan JE and McCarty OJT: Rho GTPase regulation of reactive oxygen species generation and signaling in platelet function and disease. Small GTPases. 12:440–457. 2021. View Article : Google Scholar : PubMed/NCBI

37 

Wang L, Wu Y, Zhou J, Ahmad SS, Mutus B, Garbi N, Hämmerling G, Liu J and Essex DW: Platelet-derived ERp57 mediates platelet incorporation into a growing thrombus by regulation of the αIIbβ3 integrin. Blood. 122:3642–3650. 2013. View Article : Google Scholar : PubMed/NCBI

38 

Huang J, Li X, Shi X, Zhu M, Wang J, Huang S, Huang X, Wang H, Li L, Deng H, et al: Platelet integrin αIIbβ3: Signal transduction, regulation, and its therapeutic targeting. J Hematol Oncol. 12:262019. View Article : Google Scholar

39 

Rigg RA, Healy LD, Nowak MS, Mallet J, Thierheimer ML, Pang J, McCarty OJ and Aslan JE: Heat shock protein 70 regulates platelet integrin activation, granule secretion and aggregation. Am J Physiol Cell Physiol. 310:C568–C575. 2016. View Article : Google Scholar : PubMed/NCBI

40 

De Maio A: Extracellular Hsp70: Export and function. Curr Protein Pept Sci. 15:225–231. 2014. View Article : Google Scholar : PubMed/NCBI

41 

Krause M, Heck TG, Bittencourt A, Scomazzon SP, Newsholme P, Curi R and Homem de Bittencourt PI Jr: The chaperone balance hypothesis: The importance of the extracellular to intracellular HSP70 ratio to inflammation-driven type 2 diabetes, the effect of exercise, and the implications for clinical management. Mediators Inflamm. 2015:2492052015. View Article : Google Scholar : PubMed/NCBI

42 

Jackson JW, Rivera-Marquez GM, Beebe K, Tran AD, Trepel JB, Gestwicki JE, Blagg BSJ, Ohkubo S and Neckers LM: Pharmacologic dissection of the overlapping impact of heat shock protein family members on platelet function. J Thromb Haemost. 18:1197–1209. 2020. View Article : Google Scholar : PubMed/NCBI

43 

Blagonravov ML, Sklifasovskaya AP, Korshunova AY, Azova MM and Kurlaeva AO: Heat shock protein HSP60 in left ventricular cardiomyocytes of hypertensive rats with and without insulin-dependent diabetes mellitus. Bull Exp Biol Med. 170:10–14. 2020. View Article : Google Scholar : PubMed/NCBI

44 

Henstridge DC, Whitham M and Febbraio MA: Chaperoning to the metabolic party: The emerging therapeutic role of heat-shock proteins in obesity and type 2 diabetes. Mol Metab. 3:781–793. 2014. View Article : Google Scholar : PubMed/NCBI

45 

Archer AE, Von Schulze AT and Geiger PC: Exercise, heat shock proteins and insulin resistance. Philos Trans R Soc Lond B Biol Sci. 373:201605292018. View Article : Google Scholar

46 

Drew BG, Ribas V, Le JA, Henstridge DC, Phun J, Zhou Z, Soleymani T, Daraei P, Sitz D, Vergnes L, et al: HSP72 is a mitochondrial stress sensor critical for Parkin action, oxidative metabolism, and insulin sensitivity in skeletal muscle. Diabetes. 63:1488–1505. 2014. View Article : Google Scholar : PubMed/NCBI

47 

Kitano S, Kondo T, Matsuyama R, Ono K, Goto R, Takaki Y, Hanatani S, Sakaguchi M, Igata M, Kawashima J, et al: Impact of hepatic HSP72 on insulin signaling. Am J Physiol Endocrinol Metab. 316:E305–E318. 2019. View Article : Google Scholar

48 

Xu L, Ma X, Bagattin A and Mueller E: The transcriptional coactivator PGC1α protects against hyperthermic stress via cooperation with the heat shock factor HSF1. Cell Death Dis. 7:e21022016. View Article : Google Scholar

49 

Jornayvaz FR and Shulman GI: Regulation of mitochondrial biogenesis. Essays Biochem. 47:69–84. 2010. View Article : Google Scholar : PubMed/NCBI

50 

Charos AE, Reed BD, Raha D, Szekely AM, Weissman SM and Snyder M: A highly integrated and complex PPARGC1A transcription factor binding network in HepG2 cells. Genome Res. 22:1668–1679. 2012. View Article : Google Scholar : PubMed/NCBI

51 

Ma X, Xu L, Alberobello AT, Gavrilova O, Bagattin A, Skarulis M, Liu J, Finkel T and Mueller E: Celastrol protects against obesity and metabolic dysfunction through activation of a HSF1-PGC1α transcriptional axis. Cell Metab. 22:695–708. 2015. View Article : Google Scholar : PubMed/NCBI

52 

Dang X, Du G, Hu W, Ma L, Wang P and Li Y: Peroxisome proliferator-activated receptor gamma coactivator-1α/HSF1 axis effectively alleviates lipopolysaccharide-induced acute lung injury via suppressing oxidative stress and inflammatory response. J Cell Biochem. 120:544–551. 2019. View Article : Google Scholar

53 

Meyer BA and Doroudgar S: ER Stress-induced secretion of proteins and their extracellular functions in the heart. Cells. 9:20662020. View Article : Google Scholar : PubMed/NCBI

54 

García R, Merino D, Gómez JM, Nistal JF, Hurlé MA, Cortajarena AL and Villar AV: Extracellular heat shock protein 90 binding to TGFβ receptor I participates in TGFβ-mediated collagen production in myocardial fibroblasts. Cell Signal. 28:1563–1579. 2016. View Article : Google Scholar

55 

Shi C, Ulke-Lemée A, Deng J, Batulan Z and O'Brien ER: Characterization of heat shock protein 27 in extracellular vesicles: A potential anti-inflammatory therapy. FASEB J. 33:1617–1630. 2019. View Article : Google Scholar

56 

Liu P, Bao HY, Jin CC, Zhou JC, Hua F, Li K, Lv XX, Cui B, Hu ZW and Zhang XW: Targeting extracellular heat shock protein 70 ameliorates doxorubicin-induced heart failure through resolution of toll-like receptor 2-mediated myocardial inflammation. J Am Heart Assoc. 8:e0123382019. View Article : Google Scholar : PubMed/NCBI

57 

Jan RL, Yang SC, Liu YC, Yang RC, Tsai SP, Huang SE, Yeh JL and Hsu JH: Extracellular heat shock protein HSC70 protects against lipopolysaccharide-induced hypertrophic responses in rat cardiomyocytes. Biomed Pharmacother. 128:1103702020. View Article : Google Scholar : PubMed/NCBI

58 

Zhang X, Xu Z, Zhou L, Chen Y, He M, Cheng L, Hu FB, Tanguay RM and Wu T: Plasma levels of Hsp70 and anti-Hsp70 antibody predict risk of acute coronary syndrome. Cell Stress Chaperones. 15:675–686. 2010. View Article : Google Scholar : PubMed/NCBI

59 

Jenei ZM, Gombos T, Förhécz Z, Pozsonyi Z, Karádi I, Jánoskuti L and Prohászka Z: Elevated extracellular HSP70 (HSPA1A) level as an independent prognostic marker of mortality in patients with heart failure. Cell Stress Chaperones. 18:809–813. 2013. View Article : Google Scholar : PubMed/NCBI

60 

Song YJ, Zhong CB and Wang XB: Heat shock protein 70: A promising therapeutic target for myocardial ischemia-reperfusion injury. J Cell Physiol. 234:1190–1207. 2019. View Article : Google Scholar

61 

Yang J, Yu XF, Li YY, Xue FT and Zhang S: Decreased HSP70 expression on serum exosomes contributes to cardiac fibrosis during senescence. Eur Rev Med Pharmacol Sci. 23:3993–4001. 2019.PubMed/NCBI

62 

Yoon S, Kim M, Min HK, Lee YU, Kwon DH, Lee M, Lee S, Kook T, Joung H, Nam KI, et al: Inhibition of heat shock protein 70 blocks the development of cardiac hypertrophy by modulating the phosphorylation of histone deacetylase 2. Cardiovasc Res. 115:1850–1860. 2019. View Article : Google Scholar : PubMed/NCBI

63 

Rodriguez-Iturbe B, Johnson RJ, Sanchez-Lozada LG and Pons H: HSP70 and primary arterial hypertension. Biomolecules. 13:2722023. View Article : Google Scholar : PubMed/NCBI

64 

Mathur S, Walley KR, Wang Y, Indrambarya T and Boyd JH: Extracellular heat shock protein 70 induces cardiomyocyte inflammation and contractile dysfunction via TLR2. Circ J. 75:2445–2452. 2011. View Article : Google Scholar : PubMed/NCBI

65 

Birmpilis AI, Paschalis A, Mourkakis A, Christodoulou P, Kostopoulos IV, Antimissari E, Terzoudi G, Georgakilas AG, Armpilia C, Papageorgis P, et al: Immunogenic cell death, DAMPs and prothymosin α as a putative anticancer immune response biomarker. Cells. 11:14152022. View Article : Google Scholar

66 

Bacmeister L, Schwarzl M, Warnke S, Stoffers B, Blankenberg S, Westermann D and Lindner D: Inflammation and fibrosis in murine models of heart failure. Basic Res Cardiol. 114:192019. View Article : Google Scholar : PubMed/NCBI

67 

Shah AK, Bhullar SK, Elimban V and Dhalla NS: Oxidative stress as a mechanism for functional alterations in cardiac hypertrophy and heart failure. Antioxidants (Basel). 10:9312021. View Article : Google Scholar : PubMed/NCBI

68 

Kruszewska J, Cudnoch-Jedrzejewska A and Czarzasta K: Remodeling and fibrosis of the cardiac muscle in the course of obesity-pathogenesis and involvement of the extracellular matrix. Int J Mol Sci. 23:41952022. View Article : Google Scholar : PubMed/NCBI

69 

Khalil H, Kanisicak O, Prasad V, Correll RN, Fu X, Schips T, Vagnozzi RJ, Liu R, Huynh T, Lee SJ, et al: Fibroblast-specific TGF-β-Smad2/3 signaling underlies cardiac fibrosis. J Clin Invest. 127:3770–3783. 2017. View Article : Google Scholar : PubMed/NCBI

70 

Tian J, Zhang M, Suo M, Liu D, Wang X, Liu M, Pan J, Jin T and An F: Dapagliflozin alleviates cardiac fibrosis through suppressing EndMT and fibroblast activation via AMPKα/TGF-β/Smad signalling in type 2 diabetic rats. J Cell Mol Med. 25:7642–7659. 2021. View Article : Google Scholar : PubMed/NCBI

71 

Ko T, Nomura S, Yamada S, Fujita K, Fujita T, Satoh M, Oka C, Katoh M, Ito M, Katagiri M, et al: Cardiac fibroblasts regulate the development of heart failure via Htra3-TGF-β-IGFBP7 axis. Nat Commun. 13:32752022. View Article : Google Scholar

72 

Cáceres RA, Chavez T, Maestro D, Palanca AR, Bolado P, Madrazo F, Aires A, Cortajarena AL and Villar AV: Reduction of cardiac TGFβ-mediated profibrotic events by inhibition of Hsp90 with engineered protein. J Mol Cell Cardiol. 123:75–87. 2018. View Article : Google Scholar

73 

Zhang X, Zhang Y, Miao Q, Shi Z, Hu L, Liu S, Gao J, Zhao S, Chen H, Huang Z, et al: Inhibition of HSP90 S-nitrosylation alleviates cardiac fibrosis via TGFβ/SMAD3 signalling pathway. Br J Pharmacol. 178:4608–4625. 2021. View Article : Google Scholar : PubMed/NCBI

74 

Zhong W, Chen W, Liu Y, Zhang J, Lu Y, Wan X, Qiao Y, Huang H, Zeng Z, Li W, et al: Extracellular HSP90α promotes cellular senescence by modulating TGF-β signaling in pulmonary fibrosis. FASEB J. 36:e224752022. View Article : Google Scholar

75 

Christians ES, Ishiwata T and Benjamin IJ: Small heat shock proteins in redox metabolism: Implications for cardiovascular diseases. Int J Biochem Cell Biol. 44:1632–1645. 2012. View Article : Google Scholar : PubMed/NCBI

76 

Collier MP and Benesch JLP: Small heat-shock proteins and their role in mechanical stress. Cell Stress Chaperones. 25:601–613. 2020. View Article : Google Scholar : PubMed/NCBI

77 

Nguyen VC, Deck CA and Pamenter ME: Naked mole-rats reduce the expression of ATP-dependent but not ATP-independent heat shock proteins in acute hypoxia. J Exp Biol. 222(Pt 22): jeb2112432019. View Article : Google Scholar : PubMed/NCBI

78 

Janowska MK, Baughman HER, Woods CN and Klevit RE: Mechanisms of small heat shock proteins. Cold Spring Harb Perspect Biol. 11:a0340252019. View Article : Google Scholar : PubMed/NCBI

79 

Alagar Boopathy LR, Jacob-Tomas S, Alecki C and Vera M: Mechanisms tailoring the expression of heat shock proteins to proteostasis challenges. J Biol Chem. 298:1017962022. View Article : Google Scholar : PubMed/NCBI

80 

Carver JA, Ecroyd H, Truscott RJW, Thorn DC and Holt C: Proteostasis and the regulation of intra- and extracellular protein aggregation by ATP-independent molecular chaperones: Lens α-crystallins and milk caseins. Acc Chem Res. 51:745–752. 2018. View Article : Google Scholar : PubMed/NCBI

81 

Izumi M: Heat shock proteins support refolding and shredding of misfolded proteins. Plant Physiol. 180:1777–1778. 2019. View Article : Google Scholar : PubMed/NCBI

82 

Choudhary D, Mediani L, Carra S and Cecconi C: Studying heat shock proteins through single-molecule mechanical manipulation. Cell Stress Chaperones. 25:615–628. 2020. View Article : Google Scholar : PubMed/NCBI

83 

Dokladny K, Myers OB and Moseley PL: Heat shock response and autophagy-cooperation and control. Autophagy. 11:200–213. 2015. View Article : Google Scholar :

84 

Shan Q, Ma F, Wei J, Li H, Ma H and Sun P: Physiological functions of heat shock proteins. Curr Protein Pept Sci. 21:751–760. 2020. View Article : Google Scholar

85 

Hosaka Y, Araya J, Fujita Y and Kuwano K: Role of chaperone-mediated autophagy in the pathophysiology including pulmonary disorders. Inflamm Regen. 41:292021. View Article : Google Scholar : PubMed/NCBI

86 

Wick G, Jakic B, Buszko M, Wick MC and Grundtman C: The role of heat shock proteins in atherosclerosis. Nat Rev Cardiol. 11:516–529. 2014. View Article : Google Scholar : PubMed/NCBI

87 

Bakthisaran R, Tangirala R and Rao ChM: Small heat shock proteins: Role in cellular functions and pathology. Biochim Biophys Acta. 1854:291–319. 2015. View Article : Google Scholar : PubMed/NCBI

88 

Hashikawa N, Ido M, Morita Y and Hashikawa-Hobara N: Effects from the induction of heat shock proteins in a murine model due to progression of aortic atherosclerosis. Sci Rep. 11:70252021. View Article : Google Scholar : PubMed/NCBI

89 

Cuerrier CM, Chen YX, Tremblay D, Rayner K, McNulty M, Zhao X, Kennedy CR, de BelleRoche J, Pelling AE and O'Brien ER: Chronic over-expression of heat shock protein 27 attenuates atherogenesis and enhances plaque remodeling: A combined histological and mechanical assessment of aortic lesions. PLoS One. 8:e558672013. View Article : Google Scholar : PubMed/NCBI

90 

Liu A, Ming JY, Fiskesund R, Ninio E, Karabina SA, Bergmark C, Frostegård AG and Frostegård J: Induction of dendritic cell-mediated T-cell activation by modified but not native low-density lipoprotein in humans and inhibition by annexin a5: Involvement of heat shock proteins. Arterioscler Thromb Vasc Biol. 35:197–205. 2015. View Article : Google Scholar

91 

Gong R, Li XY, Chen HJ, Xu CC, Fang HY, Xiang J and Wu YQ: Role of heat shock protein 22 in the protective effect of geranylgeranylacetone in response to oxidized-LDL. Drug Des Devel Ther. 13:2619–2632. 2019. View Article : Google Scholar : PubMed/NCBI

92 

Nahomi RB, Palmer A, Green KM, Fort PE and Nagaraj RH: Pro-inflammatory cytokines downregulate Hsp27 and cause apoptosis of human retinal capillary endothelial cells. Biochim Biophys Acta. 1842:164–174. 2014. View Article : Google Scholar :

93 

Batulan Z, Pulakazhi Venu VK, Li Y, Koumbadinga G, Alvarez-Olmedo DG, Shi C and O'Brien ER: Extracellular release and signaling by heat shock protein 27: Role in modifying vascular inflammation. Front Immunol. 7:2852016. View Article : Google Scholar : PubMed/NCBI

94 

Zhou XY, Sun JY, Wang WQ, Li SX, Li HX, Yang HJ, Yang MF, Yuan H, Zhang ZY, Sun BL and Han JX: TAT-HSP27 Peptide improves neurologic deficits via reducing apoptosis after experimental subarachnoid hemorrhage. Front Cell Neurosci. 16:8786732022. View Article : Google Scholar : PubMed/NCBI

95 

Jin C, Cleveland JC, Ao L, Li J, Zeng Q, Fullerton DA and Meng X: Human myocardium releases heat shock protein 27 (HSP27) after global ischemia: The proinflammatory effect of extracellular HSP27 through toll-like receptor (TLR)-2 and TLR4. Mol Med. 20:280–289. 2014. View Article : Google Scholar : PubMed/NCBI

96 

Inia JA and O'Brien ER: Role of Heat Shock Protein 27 in Modulating Atherosclerotic Inflammation. J Cardiovasc Transl Res. 14:3–12. 2021. View Article : Google Scholar

97 

Forouzanfar F, Butler AE, Banach M, Barreto GE and Sahbekar A: Modulation of heat shock proteins by statins. Pharmacol Res. 134:134–144. 2018. View Article : Google Scholar : PubMed/NCBI

98 

Sklifasovskaya AP and Blagonravov ML: Small heat shock proteins HSP10 and HSP27 in the left ventricular myocardium in rats with arterial hypertension and insulin-dependent diabetes mellitus. Bull Exp Biol Med. 170:699–705. 2021. View Article : Google Scholar : PubMed/NCBI

99 

Sada K, Nishikawa T, Kukidome D, Yoshinaga T, Kajihara N, Sonoda K, Senokuchi T, Motoshima H, Matsumura T and Araki E: Hyperglycemia induces cellular hypoxia through production of mitochondrial ROS followed by suppression of aquaporin-1. PLoS One. 11:e01586192016. View Article : Google Scholar : PubMed/NCBI

100 

Yu L, Chen S, Liang Q, Huang C, Zhang W, Hu L, Yu Y, Liu L, Cheng X and Bao H: Rosiglitazone reduces diabetes angiopathy by inhibiting mitochondrial dysfunction dependent on regulating HSP22 expression. iScience. 26:1061942023. View Article : Google Scholar : PubMed/NCBI

101 

Yu L, Liang Q, Zhang W, Liao M, Wen M, Zhan B, Bao H and Cheng X: HSP22 suppresses diabetes-induced endothelial injury by inhibiting mitochondrial reactive oxygen species formation. Redox Biol. 21:1010952019. View Article : Google Scholar : PubMed/NCBI

102 

Li X, Fang P, Yang WY, Chan K, Lavallee M, Xu K, Gao T, Wang H and Yang X: Mitochondrial ROS, uncoupled from ATP synthesis, determine endothelial activation for both physiological recruitment of patrolling cells and pathological recruitment of inflammatory cells. Can J Physiol Pharmacol. 95:247–252. 2017. View Article : Google Scholar

103 

Fang H, Hu N, Zhao Q, Wang B, Zhou H, Fu Q, Shen L, Chen X, Shen F and Lyu J: mtDNA haplogroup N9a increases the risk of type 2 diabetes by altering mitochondrial function and intracellular mitochondrial signals. Diabetes. 67:1441–1453. 2018. View Article : Google Scholar : PubMed/NCBI

104 

Rodríguez ME, Cogno IS, Milla Sanabria LS, Morán YS and Rivarola VA: Heat shock proteins in the context of photodynamic therapy: Autophagy, apoptosis and immunogenic cell death. Photochem Photobiol Sci. 15:1090–1102. 2016. View Article : Google Scholar : PubMed/NCBI

105 

Penke B, Bogár F, Crul T, Sántha M, Tóth ME and Vígh L: Heat shock proteins and autophagy pathways in neuroprotection: From molecular bases to pharmacological interventions. Int J Mol Sci. 19:3252018. View Article : Google Scholar : PubMed/NCBI

106 

Kanugovi Vijayavittal A, Kumar P, Sugunan S, Joseph C, Devaki B, Paithankar K and Amere Subbarao S: Heat shock transcription factor HSF2 modulates the autophagy response through the BTG2-SOD2 axis. Biochem Biophys Res Commun. 600:44–50. 2022. View Article : Google Scholar : PubMed/NCBI

107 

Cuervo AM and Wong E: Chaperone-mediated autophagy: Roles in disease and aging. Cell Res. 24:92–104. 2014. View Article : Google Scholar :

108 

Wu Z, Geng Y, Lu X, Shi Y, Wu G, Zhang M, Shan B, Pan H and Yuan J: Chaperone-mediated autophagy is involved in the execution of ferroptosis. Proc Natl Acad Sci USA. 116:2996–3005. 2019. View Article : Google Scholar : PubMed/NCBI

109 

Hale BJ, Hager CL, Seibert JT, Selsby JT, Baumgard LH, Keating AF and Ross JW: Heat stress induces autophagy in pig ovaries during follicular development. Biol Reprod. 97:426–437. 2017. View Article : Google Scholar : PubMed/NCBI

110 

Ganesan S, Pearce SC, Gabler NK, Baumgard LH, Rhoads RP and Selsby JT: Short-term heat stress results in increased apoptotic signaling and autophagy in oxidative skeletal muscle in Sus scrofa. J Therm Biol. 72:73–80. 2018. View Article : Google Scholar : PubMed/NCBI

111 

Roths M, Freestone AD, Rudolph TE, Michael A, Baumgard LH and Selsby JT: Environment-induced heat stress causes structural and biochemical changes in the heart. J Therm Biol. 113:1034922023. View Article : Google Scholar : PubMed/NCBI

112 

Li DL, Wang ZV, Ding G, Tan W, Luo X, Criollo A, Xie M, Jiang N, May H, Kyrychenko V, et al: Doxorubicin blocks cardiomyocyte autophagic flux by inhibiting lysosome acidification. Circulation. 26(133): 1668–1687. 2016. View Article : Google Scholar

113 

Packer M: Role of impaired nutrient and oxygen deprivation signaling and deficient autophagic flux in diabetic CKD development: Implications for understanding the effects of sodium-glucose cotransporter 2-inhibitors. J Am Soc Nephrol. 31:907–919. 2020. View Article : Google Scholar : PubMed/NCBI

114 

Gu S, Tan J, Li Q, Liu S, Ma J, Zheng Y, Liu J, Bi W, Sha P, Li X, et al: Downregulation of LAPTM4B contributes to the impairment of the autophagic flux via unopposed activation of mTORC1 signaling during myocardial ischemia/reperfusion injury. Circ Res. 127:e148–e165. 2020. View Article : Google Scholar : PubMed/NCBI

115 

Sciarretta S, Maejima Y, Zablocki D and Sadoshima J: The role of autophagy in the heart. Annu Rev Physiol. 80:1–26. 2018. View Article : Google Scholar

116 

Lavandero S, Troncoso R, Rothermel BA, Martinet W, Sadoshima J and Hill JA: Cardiovascular autophagy: Concepts, controversies, and perspectives. Autophagy. 9:1455–1466. 2013. View Article : Google Scholar : PubMed/NCBI

117 

Ott C, Jung T, Brix S, John C, Betz IR, Foryst-Ludwig A, Deubel S, Kuebler WM, Grune T, Kintscher U and Grune J: Hypertrophy-reduced autophagy causes cardiac dysfunction by directly impacting cardiomyocyte contractility. Cells. 10:8052021. View Article : Google Scholar : PubMed/NCBI

118 

Zhang Y, Liu D, Hu H, Zhang P, Xie R and Cui W: HIF-1α/BNIP3 signaling pathway-induced-autophagy plays protective role during myocardial ischemia-reperfusion injury. Biomed. Pharmacother. 120:1094642019. View Article : Google Scholar

119 

Liu W, Chen C, Gu X, Zhang L, Mao X, Chen Z and Tao L: AM1241 alleviates myocardial ischemia-reperfusion injury in rats by enhancing Pink1/Parkin-mediated autophagy. Life Sci. 272:1192282021. View Article : Google Scholar : PubMed/NCBI

120 

Sui Z, Wang MM, Xing Y, Qi J and Wang W: Targeting MCOLN1/TRPML1 channels to protect against ischemia-reperfusion injury by restoring the inhibited autophagic flux in cardiomyocytes. Autophagy. 18:3053–3055. 2022. View Article : Google Scholar : PubMed/NCBI

121 

Liu L, Jin X, Hu CF, Li R, Zhou Z and Shen CX: Exosomes derived from mesenchymal stem cells rescue myocardial ischaemia/reperfusion injury by inducing cardiomyocyte autophagy via AMPK and Akt pathways. Cell Physiol Biochem. 43:52–68. 2017. View Article : Google Scholar : PubMed/NCBI

122 

Xiang M, Lu Y, Xin L, Gao J, Shang C, Jiang Z, Lin H, Fang X, Qu Y, Wang Y, et al: Role of oxidative stress in reperfusion following myocardial ischemia and its treatments. Oxid Med Cell Longev. 2021:66140092021. View Article : Google Scholar : PubMed/NCBI

123 

Xing Y, Sui Z, Liu Y, Wang MM, Wei X, Lu Q, Wang X, Liu N, Lu C, Chen R, et al: Blunting TRPML1 channels protects myocardial ischemia/reperfusion injury by restoring impaired cardiomyocyte autophagy. Basic Res Cardiol. 117:202022. View Article : Google Scholar : PubMed/NCBI

124 

Kim YC and Guan KL: mTOR: A pharmacologic target for autophagy regulation. J Clin Invest. 125:25–32. 2015. View Article : Google Scholar : PubMed/NCBI

125 

Wang Y and Zhang H: Regulation of autophagy by mTOR signaling pathway. Adv Exp Med Biol. 1206:67–83. 2019. View Article : Google Scholar : PubMed/NCBI

126 

Al-Bari MAA and Xu P: Molecular regulation of autophagy machinery by mTOR-dependent and -independent pathways. Ann N Y Acad Sci. 1467:3–20. 2020. View Article : Google Scholar : PubMed/NCBI

127 

Liu GS, Zhu H, Cai WF, Wang X, Jiang M, Essandoh K, Vafiadaki E, Haghighi K, Lam CK, Gardner G, et al: Regulation of BECN1-mediated autophagy by HSPB6: Insights from a human HSPB6S10F mutant. Autophagy. 14:80–97. 2018. View Article : Google Scholar :

128 

Nicolaou P, Knöll R, Haghighi K, Fan GC, Dorn GW II, Hasenfub G and Kranias EG: Human mutation in the anti-apoptotic heat shock protein 20 abrogates its cardioprotective effects. J Biol Chem. 283:33465–33471. 2008. View Article : Google Scholar : PubMed/NCBI

129 

Shatov VM and Gusev NB: Physico-chemical properties of two point mutants of small heat shock protein HspB6 (Hsp20) with abrogated cardioprotection. Biochimie. 174:126–135. 2020. View Article : Google Scholar : PubMed/NCBI

130 

Lavandero S, Chiong M, Rothermel BA and Hill JA: Autophagy in cardiovascular biology. J Clin Invest. 125:55–64. 2015. View Article : Google Scholar : PubMed/NCBI

131 

Parzych KR and Klionsky DJ: An overview of autophagy: Morphology, mechanism, and regulation. Antioxid Redox Signal. 20:460–473. 2014. View Article : Google Scholar :

132 

Cao W, Li J, Yang K and Cao D: An overview of autophagy: Mechanism, regulation and research progress. Bull Cancer. 108:304–322. 2021. View Article : Google Scholar : PubMed/NCBI

133 

Zhou Y, Manghwar H, Hu W and Liu F: Degradation mechanism of autophagy-related proteins and research progress. Int J Mol Sci. 23:73012022. View Article : Google Scholar : PubMed/NCBI

134 

Li W, He P, Huang Y, Li YF, Lu J, Li M, Kurihara H, Luo Z, Meng T, Onishi M, et al: Selective autophagy of intracellular organelles: Recent research advances. Theranostics. 11:222–256. 2021. View Article : Google Scholar : PubMed/NCBI

135 

Li Y, Li S and Wu H: Ubiquitination-proteasome system (UPS) and autophagy two main protein degradation machineries in response to cell stress. Cells. 11:8512022. View Article : Google Scholar : PubMed/NCBI

136 

Popov SV, Mukhomedzyanov AV, Voronkov NS, Derkachev IA, Boshchenko AA, Fu F, Sufianova GZ, Khlestkina MS and Maslov LN: Regulation of autophagy of the heart in ischemia and reperfusion. Apoptosis. 28:55–80. 2023. View Article : Google Scholar

137 

Dong Y, Chen H, Gao J, Liu Y, Li J and Wang J: Molecular machinery and interplay of apoptosis and autophagy in coronary heart disease. J Mol Cell Cardiol. 136:27–41. 2019. View Article : Google Scholar : PubMed/NCBI

138 

Denton D and Kumar S: Autophagy-dependent cell death. Cell Death Differ. 26:605–616. 2019. View Article : Google Scholar :

139 

Mahapatra KK, Mishra SR, Behera BP, Patil S, Gewirtz DA and Bhutia SK: The lysosome as an imperative regulator of autophagy and cell death. Cell Mol. Life Sci. 78:7435–7449. 2021. View Article : Google Scholar : PubMed/NCBI

140 

Xu HD and Qin ZH: Beclin 1, Bcl-2 and Autophagy. Adv Exp Med Biol. 1206:109–126. 2019. View Article : Google Scholar : PubMed/NCBI

141 

Liu J, Liu W and Yang H: Balancing apoptosis and autophagy for Parkinson's disease therapy: Targeting BCL-2. ACS Chem. Neurosci. 10:792–802. 2019.

142 

Blagonravov ML, Sklifasovskaya AP, Demurov EA and Karimov AA: Beclin-1-dependent autophagy of left ventricular cardiomyocytes in SHR and Wistar-Kyoto rats with type 1 diabetes mellitus. Bull Exp Biol Med. 171:23–27. 2021. View Article : Google Scholar : PubMed/NCBI

143 

Sklifasovskaya AP, Blagonravov ML, Ryabinina AY, Azova MM and Goryachev VA: Expression of Bax and Bcl-2 Proteins in Left-Ventricular Cardiomyocytes in Wistar-Kyoto and SHR Rats with Insulin-Dependent Diabetes Mellitus. Bull Exp Biol Med. 171:576–581. 2021. View Article : Google Scholar : PubMed/NCBI

144 

Van Opdenbosch N and Lamkanfi M: Caspases in cell death, inflammation, and disease. Immunity. 50:1352–1364. 2019. View Article : Google Scholar : PubMed/NCBI

145 

Araya LE, Soni IV, Hardy JA and Julien O: Deorphanizing caspase-3 and caspase-9 substrates in and out of apoptosis with deep substrate profiling. ACS Chem Biol. 16:2280–2296. 2021. View Article : Google Scholar : PubMed/NCBI

146 

Green DR: Caspase activation and inhibition. Cold Spring Harb Perspect Biol. 14:a0410202022. View Article : Google Scholar : PubMed/NCBI

147 

Kashyap D, Garg VK and Goel N: Intrinsic and extrinsic pathways of apoptosis: Role in cancer development and prognosis. Adv Protein Chem Struct Biol. 125:73–120. 2021. View Article : Google Scholar : PubMed/NCBI

148 

Lossi L: The concept of intrinsic versus extrinsic apoptosis. Biochem J. 479:357–384. 2022. View Article : Google Scholar : PubMed/NCBI

149 

Tang D, Kang R, Berghe TV, Vandenabeele P and Kroemer G: The molecular machinery of regulated cell death. Cell Res. 29:347–364. 2019. View Article : Google Scholar : PubMed/NCBI

150 

Obeng E: Apoptosis (programmed cell death) and its signals-A review. Braz J Biol. 81:1133–1143. 2021. View Article : Google Scholar

151 

Kennedy D, Jäger R, Mosser DD and Samali A: Regulation of apoptosis by heat shock proteins. IUBMB Life. 66:327–338. 2014. View Article : Google Scholar : PubMed/NCBI

152 

Leung AM, Redlak MJ and Miller TA: Role of heat shock proteins in oxygen radical-induced gastric apoptosis. J Surg Res. 193:135–144. 2015. View Article : Google Scholar

153 

Yu Y, Hu LL, Liu L, Yu LL, Li JP, Rao JA, Zhu LJ, Bao HH and Cheng XS: Hsp22 ameliorates lipopolysaccharide-induced myocardial injury by inhibiting inflammation, oxidative stress, and apoptosis. Bioengineered. 12:12544–12554. 2021. View Article : Google Scholar : PubMed/NCBI

154 

Ruan L, Zhou C, Jin E, Kucharavy A, Zhang Y, Wen Z, Florens L and Li R: Cytosolic proteostasis through importing of misfolded proteins into mitochondria. Nature. 543:443–446. 2017. View Article : Google Scholar : PubMed/NCBI

155 

Koike N, Hatano Y and Ushimaru T: Heat shock transcriptional factor mediates mitochondrial unfolded protein response. Curr Genet. 64:907–917. 2018. View Article : Google Scholar : PubMed/NCBI

156 

Verma A, Sumi S and Seervi M: Heat shock proteins-driven stress granule dynamics: Yet another avenue for cell survival. Apoptosis. 26:371–384. 2021. View Article : Google Scholar : PubMed/NCBI

157 

Liyanagamage DSNK and Martinus RD: Role of mitochondrial stress protein HSP60 in diabetes-induced neuroinflammation. Mediators Inflamm. 2020:80735162020. View Article : Google Scholar : PubMed/NCBI

158 

Kumar R, Chaudhary AK, Woytash J, Inigo JR, Gokhale AA, Bshara W, Attwood K, Wang J, Spernyak JA, Rath E, et al: A mitochondrial unfolded protein response inhibitor suppresses prostate cancer growth in mice via HSP60. J Clin Invest. 132:e1499062022. View Article : Google Scholar : PubMed/NCBI

159 

Duan Y, Tang H, Mitchell-Silbaugh K, Fang X, Han Z and Ouyang K: Heat shock protein 60 in cardiovascular physiology and diseases. Front Mol Biosci. 7:732020. View Article : Google Scholar : PubMed/NCBI

160 

Song E, Tang S, Xu J, Yin B, Bao E and Hartung J: Lenti-siRNA Hsp60 promote bax in mitochondria and induces apoptosis during heat stress. Biochem Biophys Res Commun. 481:125–131. 2016. View Article : Google Scholar : PubMed/NCBI

161 

Tian X, Zhao L, Song X, Yan Y, Liu N, Li T, Yan B and Liu B: HSP27 inhibits homocysteine-induced endothelial apoptosis by modulation of ROS production and mitochondrial caspase-dependent apoptotic pathway. Biomed Res Int. 2016:48478742016. View Article : Google Scholar : PubMed/NCBI

162 

Kennedy D, Mnich K, Oommen D, Chakravarthy R, Almeida-Souza L, Krols M, Saveljeva S, Doyle K, Gupta S, Timmerman V, et al: HSPB1 facilitates ERK-mediated phosphorylation and degradation of BIM to attenuate endoplasmic reticulum stress-induced apoptosis. Cell Death Dis. 8:e30262017. View Article : Google Scholar : PubMed/NCBI

163 

Önay Uçar E and Şengelen A: Resveratrol and siRNA in combination reduces Hsp27 expression and induces caspase-3 activity in human glioblastoma cells. Cell Stress Chaperones. 24:763–775. 2019. View Article : Google Scholar : PubMed/NCBI

164 

Guo S, Gao C, Xiao W, Zhang J, Qu Y, Li J and Ye F: Matrine protects cardiomyocytes from ischemia/reperfusion injury by regulating HSP70 expression via activation of the JAK2/STAT3 pathway. Shock. 50:664–670. 2018. View Article : Google Scholar : PubMed/NCBI

165 

Xin BR, Li P, Liu XL and Zhang XF: Visfatin relieves myocardial ischemia-reperfusion injury through activation of PI3K/Akt/HSP70 signaling axis. Eur Rev Med Pharmacol Sci. 24:10779–10789. 2020.PubMed/NCBI

166 

Huang C, Deng H, Zhao W and Xian L: Knockdown of miR-384-3p protects against myocardial ischemia-reperfusion injury in rats through targeting HSP70. Heart Surg Forum. 24:E143–E150. 2021. View Article : Google Scholar : PubMed/NCBI

167 

Song N, Ma J, Meng XW, Liu H, Wang H, Song SY, Chen QC, Liu HY, Zhang J, Peng K and Ji FH: Heat shock protein 70 protects the heart from ischemia/reperfusion injury through inhibition of p38 MAPK Signaling. Oxid Med Cell Longev. 2020:39086412020. View Article : Google Scholar : PubMed/NCBI

168 

Choudhury S, Bae S, Ke Q, Lee JY, Kim J and Kang PM: Mitochondria to nucleus translocation of AIF in mice lacking Hsp70 during ischemia/reperfusion. Basic Res Cardiol. 106:397–407. 2011. View Article : Google Scholar : PubMed/NCBI

169 

Zhang C, Liu X, Miao J, Wang S, Wu L, Yan D, Li J, Guo W, Wu X and Shen A: Heat shock protein 70 protects cardiomyocytes through suppressing SUMOylation and nucleus translocation of phosphorylated eukaryotic elongation factor 2 during myocardial ischemia and reperfusion. Apoptosis. 22:608–625. 2017. View Article : Google Scholar : PubMed/NCBI

170 

Sun A, Zou Y, Wang P, Xu D, Gong H, Wang S, Qin Y, Zhang P, Chen Y, Harada M, et al: Mitochondrial aldehyde dehydrogenase 2 plays protective roles in heart failure after myocardial infarction via suppression of the cytosolic JNK/p53 pathway in mice. J Am Heart Assoc. 3:e0007792014. View Article : Google Scholar : PubMed/NCBI

171 

Jenei ZM, Széplaki G, Merkely B, Karádi I, Zima E and Prohászka Z: Persistently elevated extracellular HSP70 (HSPA1A) level as an independent prognostic marker in post-cardiac-arrest patients. Cell Stress Chaperones. 18:447–454. 2013. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2023
Volume 52 Issue 5

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Sklifasovskaya AP, Blagonravov M, Ryabinina A, Goryachev V, Syatkin S, Chibisov S, Akhmetova K, Prokofiev D and Agostinelli E: The role of heat shock proteins in the pathogenesis of heart failure (Review). Int J Mol Med 52: 106, 2023
APA
Sklifasovskaya, A.P., Blagonravov, M., Ryabinina, A., Goryachev, V., Syatkin, S., Chibisov, S. ... Agostinelli, E. (2023). The role of heat shock proteins in the pathogenesis of heart failure (Review). International Journal of Molecular Medicine, 52, 106. https://doi.org/10.3892/ijmm.2023.5309
MLA
Sklifasovskaya, A. P., Blagonravov, M., Ryabinina, A., Goryachev, V., Syatkin, S., Chibisov, S., Akhmetova, K., Prokofiev, D., Agostinelli, E."The role of heat shock proteins in the pathogenesis of heart failure (Review)". International Journal of Molecular Medicine 52.5 (2023): 106.
Chicago
Sklifasovskaya, A. P., Blagonravov, M., Ryabinina, A., Goryachev, V., Syatkin, S., Chibisov, S., Akhmetova, K., Prokofiev, D., Agostinelli, E."The role of heat shock proteins in the pathogenesis of heart failure (Review)". International Journal of Molecular Medicine 52, no. 5 (2023): 106. https://doi.org/10.3892/ijmm.2023.5309