Next Article in Journal
Brusatol Inhibits Esophageal Squamous Cell Carcinoma Tumorigenesis Through Bad-Mediated Mitochondrial Apoptosis Induction and Anti-Metastasis by Targeting Akt1
Previous Article in Journal
Expression of FGF23 and α-KLOTHO in Normal Human Kidney Development and Congenital Anomalies of the Kidney and Urinary Tract (CAKUT)
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Proton-Sensing G Protein-Coupled Receptors and Their Potential Role in Exercise Regulation of Arterial Function

School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China
*
Authors to whom correspondence should be addressed.
Biomolecules 2025, 15(6), 813; https://doi.org/10.3390/biom15060813
Submission received: 8 April 2025 / Revised: 29 May 2025 / Accepted: 31 May 2025 / Published: 4 June 2025
(This article belongs to the Section Biological Factors)

Abstract

:
During periods of exercise, the primary cause of metabolic acidosis is the accumulation of lactate from anaerobic metabolism, whereas a transient increase in CO2 triggers a mild respiratory acidosis through the production of carbonic acid (H2CO3). The combined effects of these reactions result in a slight acidifying shift in arterial blood pH. Proton-sensing G protein-coupled receptors (including GPR68, GPR4, GPR132, and GPR65) represent the primary receptors within the body for detecting alterations in extracellular proton concentrations. These receptors have been demonstrated to possess potential roles in mechanosensation, intestinal inflammation, oncoimmunological interactions, hematopoiesis, as well as inflammatory and neuropathic pain. Recent studies have shown that the activation or inhibition of these receptors modulates a number of arterial functions, including angiogenesis, arterial relaxation, and arterial inflammation. It is well established that moderate exercise has a beneficial effect on the regulation of arterial function. This study examines the effect of exercise on proton concentrations in the microenvironment of the organism and its influence on proton-sensing G protein-coupled receptors located on cell membranes, as well as possible mechanisms involved in the regulation of arterial function. The objective is to present novel perspectives for the exploration of potential drug targets for the prevention and treatment of arterial dysfunction and the development of exercise regimens.

1. Introduction

Cardiovascular diseases (CVDs) are responsible for more than 17 million disease-related deaths worldwide each year, representing the leading cause of death [1,2]. Approximately 35.6 million deaths will be attributed to CVDs by 2050 [3]. Atherosclerosis represents a fundamental pathological marker for the assessment of cardiovascular well-being and is strongly associated with the incidence of CVDs and all-cause mortality. Increased arterial stiffness, which is primarily caused by structural abnormalities of the arterial wall [4], can result in an elevated left ventricular afterload through the action of artery–left ventricular coupling mechanisms. This may result in additional damage to other organs, including the heart and brain [5]. Impaired endothelium-dependent vasodilator function is regarded as a pivotal instigator of atherosclerosis [6,7]. There is a growing awareness of the potential benefits of enhancing arterial diastolic function in reducing the morbidity and mortality associated with CVD.
A substantial body of evidence has accumulated over the years, establishing the efficacy of exercise in the prevention and treatment of CVDs [8,9]. The current mechanisms by which exercise affects arterial function are largely manifested through the regulation of vascular endothelial cell (VEC) and vascular smooth muscle cell (VSMC) activity. This includes the regulation of endothelial cell integrity, which results in the improvement of selective barrier function [10], reduced adhesion to leukocytes [11], anti-thrombogenicity [12], angiogenesis, and regulation of vascular tone [13]. It also encompasses the modulation of endothelial function, including the regulation of increased flow shear stress [14], and the attenuation of inflammatory processes [15,16]. Additionally, it has been demonstrated that exercise may have a beneficial effect on atherosclerosis, including increased collagen content and decreased intercellular adhesion molecule-1 [17]. These adaptations have been associated with a reduction in both necrotic core area and plaque burden [18]. Furthermore, structural adaptations have been observed following exercise, including improvements in coronary artery size [19] and distensibility [20], increases in lumen diameter [21], and reductions in wall thickness [22].
During exercise, the body’s metabolic rate increases to meet the energy demands of working muscles, leading to enhanced production of CO2. The rise in CO2 promotes the formation of H2CO3, contributing to a mild respiratory acidosis. However, this effect is largely counteracted by compensatory hyperventilation, which rapidly eliminates excess CO2. The predominant cause of acidification in intense exercise is instead lactic acid accumulation from anaerobic metabolism, driving metabolic acidosis and a slight decrease in arterial blood pH [23,24]. VECs and VSMCs within arteries express proton-sensing G protein-coupled receptors (GPCRs), including the ovarian cancer G protein-coupled receptor (OGR1) (also known as GPR68), GPR4, G2 accumulation protein (G2A) (also known as GPR132), and T-cell death-associated gene 8 (TDAG8) (also known as GPR65) [25,26,27,28,29]. Such receptors are capable of detecting alterations in extracellular pH levels, which then trigger intracellular signaling cascades [30,31]. These receptors are expressed in diverse cells that regulate central pH homeostasis, pH sensing in the immune system, and vascular responses to pH [32]. In the cardiovascular system, the binding of excess protons in the acidic extracellular medium to receptors on the cell membrane results in alterations to receptor shape and the subsequent activation or inhibition of downstream signaling in VECs and VSMCs [33,34]. These interactions then regulate a number of processes, including angiogenesis, arterial diastole, and inflammation. The objective of this review was to examine the effect of physical activity on the concentration of protons in the microenvironment within the organism and to analyze whether these protons can act via proton-sensing GPCRs on the membranes of VECs and VSMCs, leading to improved arterial function.

2. Overview of Proton-Sensing GPCRs

2.1. Structure and Function

GPCRs, also known as 7-transmembrane structural domain receptors (7TMRs), constitute a diverse family of proteins that are encoded by the human genome. To date, more than 800 GPCRs have been identified [35]. The intracellular pH of cells is regulated within the range of 7.1 to 7.2 by the control of membrane proton pumps and transport proteins, through the utilization of pH sensors located in the cell membrane. Alterations in extracellular pH changes lead to the activation of specific acid-sensitive ion channels (ASICs), proton-sensing GPCRs, and lactate receptors, among others, which regulate cellular function [36]. Proton-sensing GPCRs are capable of detecting alterations in the concentration of H+ in the vicinity of histidine residues that are present in their extracellular domains.
Proton-sensing GPCRs are involved in a number of biological processes, including cell proliferation, metastasis, angiogenesis, apoptosis, immune cell functioning, and inflammatory responses [37,38]. Members of the pH-sensing GPCR family, including GPR68, GPR4, GPR132, and GPR65, have the capacity to detect alterations in extracellular proton concentrations [39] (see Table 1). The receptors demonstrate minimal activity at pH 7.6–7.8 and attain full activation at the slightly lower pH range of 6.4–6.8 [40]. In physiological conditions, GPR4, GPR68, and GPR132 are expressed in a ubiquitous manner. However, GPR132 shows the strongest expression in leukocytes, including T- and B-cells, neutrophils, and macrophages. In contrast, GPR65 is expressed almost exclusively in lymphoid tissue [41]. With regard to the context of arteries, proton-sensing GPCRs are predominantly expressed in VECs and VSMCs.

2.2. Signaling Pathway

GPCRs are capable of transducing signals from the external environment by activating G proteins, including Gαi/o, Gαq/11, Gαs, and G12/13. These proteins regulate the activity of specific enzymes, ion channels, and proteins involved in signaling cascades [42]. It is worth noting that proton-sensing GPCRs do not bind complex extracellular ligands; however, they are involved in heterotrimeric G protein responses when the extracellular proton concentration increases slightly [43]. In the presence of an extracellular acidic microenvironment, these receptors become active, and their activation of multiple downstream pathways plays a crucial role in cellular functions and processes.
Several models of the mechanism by which proton-sensing GPCRs recognize protons have been proposed. Proton-sensing GPCRs harbor an abundance of extracellular histidine residues that likely titrate at physiologically relevant pH levels [44]. The prevailing perspective suggests that proton-sensing GPCRs detect the acidic extracellular environment through a process involving protonation and coordination of several amino acids, including extracellular histidines and buried amino acid triads containing aspartic acid and two glutamic acids [43,44,45]. Subsequent endocytosis of proton-sensing GPCRs transports them to the acidic endosomal compartment, where there is a greater likelihood of receptor activation [45]. However, mutational studies suggest that histidines are dispensable for proton sensing, as the removal of all extracellular histidines does not abolish proton-driven activation in GPR68 [32,46]. At this juncture, the mechanisms by which protons activate proton-sensing GPCRs, whether proton-sensing GPCRs can be selectively activated in endosomal compartments, and whether they follow the tight coupling of transport and signaling described for most known GPCRs, remain to be elucidated.
Existing studies have found that GPR68 displays weak activity at pH 7.8 but is fully active at pH 6.8 [44]. In instances of extracellular acidosis, GPR68 may interact with a range of G proteins, thereby initiating signaling cascades. Upon coupling with Gαq/11 protein, it is capable of activating the phospholipase C (PLC)/Ca2+ signaling pathway [40]. Activation of the PLC/Ca2+ signaling pathway in human ovarian cancer cells has been demonstrated to inhibit cell proliferation and migration. Furthermore, it has been demonstrated to increase the expression of proteins such as extracellular matrix fibronectin, ovalbumin, and collagen type I and type IV, while also enhancing cellular adhesion [47]. Upon coupling with Gαs and G13 proteins, GPR68 can activate the Gαs/cyclic adenosine monophosphate (cAMP) and Ras homologous (Rho)/Rho-associated protein kinase (ROCK) pathways, thereby participating in cell physiological regulatory processes [48]. The coupling of GPR68 with Gαq and Gαs proteins has been demonstrated to regulate cell proliferation, migration, and adhesion [49]. In addition, GPR65 and GPR4 are capable of binding to Gαs proteins, which activates the downstream adenylate cyclase (AC)/cAMP signaling pathway and plays a role in regulating the inflammatory process [50,51]. Additionally, GPR65 has been demonstrated to interact with G12/13 proteins, which may play a role in the activation of the Rho signaling pathway and subsequently inhibit the production of pro-inflammatory cytokines and chemokines [52]. In the event of carbonic acidosis, GPR4 activates the Gαs/cAMP/exchange proteins and Gαq/PLC/Ca2+ and G13/Rho signaling pathways, which in turn increases the adhesion of VECs to leukocytes, thereby indirectly exacerbating the inflammatory response [53]. Other studies have demonstrated that GPR132 is responsive to the acidic milieu of plasma, which results in the activation of several downstream conductance pathways, including PLC/Ca2+, G13/Rho, and Ras/extracellular signal-regulated kinase (ERK). Furthermore, these pathways are also actively involved in the processes of oxidative stress and inflammation [48,54]. Collectively, proton-sensing GPCRs exert a pivotal influence on a number of cellular processes, including proliferation, migration, adhesion, and inflammatory responses (see Figure 1).

3. The Role and Mechanism of Proton-Sensing GPCRs in the Regulation of Arterial Function

Arterial dysfunction is a broad term that encompasses a range of pathological states. These include, but are not limited to, endothelial dysfunction, arterial stiffness, atherosclerosis, hypertension, and abnormal angiogenesis. The regulation of arterial function is primarily mediated by VECs and VSMCs. This section presents a synthesis of existing literature to evaluate the regulatory functions and potential mechanisms of proton-sensing GPCRs in the pathophysiology of arterial function.

3.1. GPR68/OGR1

GPR68, also known as OGR1, is located on chromosome 14q31 and serves as a sensor for alterations in extracellular hydrogen ion concentration [25]. It has been demonstrated that the expression of GPR68-dependent genes induced by extracellular acidification is significantly enhanced under hypoxic conditions [55]. GPR68 has been linked to a number of physiological processes, including renal function and bone metabolism [56,57]. A recent observation reported that a homozygous loss of function of GPR68 was described in families with amelogenesis imperfecta, which suggests that GPR68 is required for dental enamel formation [55]. In the arteries, GPR68 is predominantly expressed in endothelial cells of small-diameter vessels [25]. The results of studies conducted on GPR68 knockout mice indicate that GPR68 plays a crucial role in the vasodilation and outward remodeling of small-diameter arteries [25]. Furthermore, GPR68 plays a pivotal role in regulating inflammation, promoting cellular proliferation, facilitating migration and adhesion, as well as promoting angiogenesis [58].
Research addressing the regulation of arterial function by GPR68 has predominantly concentrated on the in vitro cellular level. In an environment with low acidity, GPR68 has the capacity to activate the ERK and mitogen-activated protein kinase (MAPK) signaling pathways, which in turn results in the production of inflammatory factors, such as interleukin-6 (IL-6) [59]. In the presence of an extracellular acidic microenvironment, GPR68 facilitates the formation of basal stress fibers comprising F-actin, thereby enhancing the function of the epithelial barrier [60]. Activation of the PLC/cyclooxygenase (COX)/prostaglandin-I-2 (PGI2) pathway was also observed, resulting in the production of PGI2 and the accumulation of cAMP in aortic smooth muscle cells (AoSMCs) [61]. Liu et al. [62] discovered that the extracellular acidification of the GPR68/Gαq/11 pathway can influence the expression of the COX-2 protein and mRNA in AoSMCs, resulting in the production of PGI2 and cAMP. These ultimately act in concert with lysophosphatidic acid (LPA)/Gαi to elicit an anti-atherosclerotic effect. The results of the cell-based experiments demonstrated that GPR68 activity increased in response to a decrease in the pH of the surrounding environment. Additionally, a medium with a pH of 6.4 was observed to inhibit the proliferation, migration, and tube-forming process of endothelial progenitor cells (EPCs). The effects of this environment on EPCs were partially reversed by the knockdown of GPR68 with siRNA [63] (see Figure 2). Therefore, GPR68 exerts regulatory functions with regard to the proliferation, differentiation, and migration of VECs and VSMCs within an acidic microenvironment. Further investigation is required to elucidate the complete role and mechanism of GPR68 in regulating arterial function at different pH values.

3.2. GPR4

The GPR4 gene is located on chromosome 19q13.3 and encodes a protein consisting of 362 amino acids. It is widely expressed in various tissues, including the vascular system, lung, liver, and intestine [30]. GPR4 is a proton-sensing receptor that exhibits low activity at a plasma pH of 7.4. However, it can be fully activated in acidic conditions, which is crucial for regulating the proliferation, migration, and angiogenesis of VECs [64]. Additionally, it has been demonstrated that GPR4 facilitates EPC-induced angiogenesis by activating the signal transducer and activator of transcription 3 (STAT3)/vascular endothelial growth factor A (VEGFA) pathway in patients with coronary artery disease [65]. In cellular experiments, the modulation of Notch receptor 1 (Notch1) expression through the use of siRNA or Notch receptor inhibitors has been demonstrated to significantly enhance GPR4-induced endothelial vasculogenesis and lymphocyte transendothelial migration in human microvessels [66]. Furthermore, GPR4 has been shown to promote angiogenesis and maintain vascular integrity and stability by increasing the expression of VEGF receptors [67]. Activation of GPR4 in the acidic microenvironment of tumors has been shown to lead to the activation of the G12/13/Rho guanosine triphosphatase (GTPase) signaling pathway, which in turn results in the formation of paracellular gaps in VECs. This process is positively correlated with the expression of VEC proliferation markers [68].
Notably, Wenzel et al. [69] discovered that GPR4 activates the Gαq/11-related pathway in VECs within a low-pH microenvironment, resulting in impaired cerebrovascular reactivity and anxiety induction. Furthermore, GPR4 activates the C/EBP homologous protein (CHOP) pathway, which ultimately results in apoptosis in HK-2 cells and human umbilical vein endothelial cells (HUVECs) [70]. With regard to the phenomenon of inflammation, a low-pH microenvironment activates the GPR4 in VECs. This activation results in the expression of stress response genes related to the endoplasmic reticulum, including CHOP, activate transcription factor 3 (ATF3), and vesicle NOD-, LRR-, and pyrin domain-containing protein 3 (NLRP3) expression. Consequently, this leads to the onset of inflammatory responses and apoptosis [71]. In a mouse model of acute hindlimb ischemia-reperfusion, GPR4 has been demonstrated to mediate a number of key processes, including tissue edema, inflammatory exudate formation, endothelial adhesion molecule expression, and leukocyte infiltration. Specific knockdown or pharmacological inhibition of GPR4 has been observed to result in attenuated tissue inflammation [26]. In an acidic environment, GPR4 plays a role in the transcription of genes associated with inflammatory mediators (such as NF-κB, NF-κB1, and NF-κB2) and adhesion molecules (such as E-selectin, vascular cell adhesion molecule-1 (VCAM-1), and intercellular adhesion molecule-1 (ICAM-1)). The inflammatory response induced by acidosis is significantly reduced by the administration of a GPR4 antagonist [72] (see Figure 2). To sum up, the activation of GPR4 in a low-pH microenvironment appears to play a role in arterial inflammation and may contribute to processes such as apoptosis and atherosclerosis. Furthermore, while the majority of current research on GPR4 is focused on VECs and the regulation of VSMCs remains uncertain, it may still have a positive impact on arterial function by regulating VEC proliferation, migration, and angiogenesis in a low-pH microenvironment. It is imperative to acknowledge the dual effect of a low pH microenvironment on the regulation of arterial function via GPR4.

3.3. GPR132/G2A

GPR132, also known as G2A, has been demonstrated to induce cell cycle arrest in the G2/M phase, delay mitotic progression, and diminish the transforming potential of BCR-ABL. It was regarded as a cell cycle regulator that inhibits cell proliferation [41]. In contrast to other members of the G-protein family, GPR132 displays only minimal proton sensitivity [73]. There is currently a debate among the scientific community regarding the role of GPR132 in the control of inflammatory processes. Wu et al. [74] observed a significant elevation in the levels of the proton-sensing receptor GPR132 and the inflammatory factor tumor necrosis factor-α (TNF-α) in the peripheral blood cells of patients diagnosed with pulmonary hypertension, in comparison to other G proteins. GPR132 has been demonstrated to exacerbate the inflammatory response by stimulating the secretion of pro-inflammatory factors, specifically IL-6 and IL-8, and promoting calcium mobilization [75]. Moreover, it has been demonstrated that this protein can induce M1-type macrophage polarization at sites of inflammation [76]. However, alternative research has indicated that GPR132 may play a role in dampening inflammatory and autoimmune responses by influencing the migration of monocytes and macrophages [77].
Furthermore, in the acidic tumor microenvironment, activated GPR132 has the capacity to promote M2-like macrophage polarization and inhibit inflammatory processes [78]. In relation to arterial stiffness, Parks et al. [79] discovered through experimentation that elevated serum levels of high-density lipoprotein (HDL) had no effect on the initial stages of atherosclerosis in GPR132-deficient LDLR KO mice. In contrast, Bolick et al. and Johnson et al. [27,80] previously conducted research on GPR132-deficient ApoE KO mice as an alternative animal model. It was observed that during the mid to late stages of atherosclerosis, there was an increase in the number of monocytes and pro-inflammatory M1-type macrophages. This influx was accompanied by enhanced adhesion of VECs, increased apoptosis, and larger atheromatous plaques. It may therefore be the case that GPR132 exerts a time-dependent influence on the development of atherosclerosis, with a reduced impact in the early stages of the disease and a manifesting of its pro-lesion properties during the intermediate and advanced stages of atherosclerosis (see Figure 2). In conclusion, while GPR132 may have a bidirectional function, primarily in macrophage-mediated pro-inflammatory and anti-inflammatory responses, there is currently a paucity of studies examining the proton sensitivity of GPR132. Additionally, the precise molecular mechanisms underlying the physiological functions of monocytes and macrophages in the regulation of atherosclerosis remain to be elucidated.

3.4. GPR65/TDAG8

GPR65, also known as TDAG8, was initially identified by Ishii et al. [81] as a proton-sensing receptor. It is located on chromosome 14q31-32.1 and exhibits a distinctive expression profile within the immune system. TDAG8 is predominantly expressed in cells of the immune system [82]. In humans, TDAG8 is predominantly expressed in peripheral blood leukocytes and lymphoid tissues, including the spleen, lymph nodes, and thymus [83], which suggests a pivotal role in innate and adaptive immune responses. Moreover, it is implicated in the accumulation of cAMP, Rho activation, and stress fiber formation. In response to extracellular acidification, TDAG8 plays a pivotal role in leukocyte migration and phagocytosis through the activation of the Gαs/AC/cAMP/PKA pathway, the G12/13 proteins, and the Rho-related pathway [81,84]. With respect to the arterial system, the primary function of this protein is to regulate the process of atherosclerosis.
In ApoE KO mice, an elevation in mRNA and protein expression levels of GPR65 was observed. Immunofluorescent staining has shown that GPR65 is predominantly distributed within proliferating cell nuclear antigen (PCNA)-positive VSMCs. It is proposed that this activation may regulate the phenotypic transformation, proliferation, and migration of VSMCs by activating the downstream cAMP/PKA pathway. This cascade subsequently exerts an influence on the pathological process of atherosclerosis [28]. It is proposed that GPR65 may serve as a pivotal regulator and therapeutic target in the context of myocardial infarction (MI). In the aftermath of MI, GPR65 prompts an uptick in lactate generated by myocardial anaerobic glycolytic metabolism and myocardial immune cell infiltration, culminating in cardiac ischemic acidosis. Mice lacking the GPR65 gene have been observed to overproduce IL-17A, which contributes to the deterioration of cardiac function following MI, resulting in a significantly lower survival rate and cardiac function compared to wild-type mice [85] (see Figure 2). Taken together, GPR65 contributes to the regulation of arterial function by mediating the proliferation, differentiation, and migration of VSMCs. A number of studies have investigated the role of GPR65 in arterial function, with a particular focus on its effects on plasma pH. The findings of these studies have consistently indicated that GPR65 has a beneficial effect on arterial function. Nevertheless, the precise mechanism by which GPR65 functions as a proton-sensing receptor in arterial function remains to be elucidated.

4. The Effects of Exercise on the Regulation of Arterial Function

Modifications in blood flow and shear stress are known to precipitate arterial remodeling, which is contingent upon the presence of a fully functional endothelium [86,87]. Additionally, VECs are instrumental in regulating vascular tone and blood pressure [88]. The hemodynamic changes that occur in response to exercise can directly or indirectly affect the diastole of arterioles, thereby prompting adaptive alterations in the arteries. These changes are mediated by the regulation of the function of both VECs and VSMCs [21,89,90,91]. The available evidence indicates that regular exercise may delay the onset of age-related arterial lesions. The impact of exercise on arterial function is subject to variation in terms of the type, intensity, and frequency of activity, and there may be heterogeneity in the effects on different age groups and populations. Research has shown that low- and moderate-intensity aerobic exercise can prevent the development of atherosclerosis in large arteries and improve endothelial function in sedentary middle-aged and older adults. Moreover, it has been demonstrated to reduce arterial stiffness and restore endothelial function [92]. It has been demonstrated that low-intensity exercise is significantly less effective than high-intensity exercise in improving exercise capacity in patients with peripheral arterial disease [93]. Additionally, research findings suggest that exercise training can reduce arterial stiffness and improve arterial compliance in the context of atherosclerosis [94,95]. Another study found that high-intensity resistance exercise resulted in a reduction in arterial compliance and an increase in arterial stiffness, whereas low-intensity resistance exercise led to a notable increase in arterial compliance and a decrease in arterial stiffness [96]. In summary, different types of exercise can induce modifications in arterial pressure, blood flow, and shear stress, which consequently lead to alterations in arterial diastolic function and stiffness (see Table 2).
In the field of research examining the impact of varying exercise durations on arterial function, acute aerobic exercise has been demonstrated to be effective in reducing central arterial stiffness, wave reflections, and hemodynamics in healthy populations [97,98,99]. Nevertheless, acute resistance exercise has been demonstrated to result in a transient increase in central artery stiffness [100]. Other studies have found that high-intensity aerobic exercise, or HIIT, three days per week for eight weeks has been found to be more beneficial in improving arterial stiffness in younger populations compared to acute aerobic or resistance training [101,102]. Long-term aerobic exercise (lasting for 12 weeks) has been shown to significantly improve atherosclerotic coronary endothelial dysfunction and reduce arterial inflammation and oxidative stress [103]. Another investigation demonstrated that long-term aerobic exercise (with an intervention period of 12–20 weeks) lessened arterial stiffness in hypertensive women [104]. A similar discovery was made in a year-long program of moderate-to-high-intensity aerobic exercise, which led to a significant decrease in carotid artery stiffness and enhancement in cerebral blood flow in patients with amnestic mild cognitive impairment [105]. Furthermore, a combination of long-term aerobic and resistance exercise (lasting between 6 and 12 months) has been demonstrated to significantly enhance arterial blood pressure and arterial function in patients with chronic kidney disease [106]. Conversely, an 8-week resistance exercise intervention had no effect on pulse wave velocity or arterial stiffness in patients with metabolic syndrome [107]. The current body of research suggests that the effect of exercise on arterial function is time-dependent. Specifically, arterial stiffness and compliance appear to be more positively impacted by prolonged exercise durations. However, it should be noted that the effects on arterial function are influenced by numerous exercise-related factors, including the intensity and nature of the exercise modality. Furthermore, the potential benefits in diverse disease states necessitate additional investigation.

5. Regulation of Arterial Function by Exercise Through Proton-Sensing GPCRs

It is established that both proton-sensing GPCRs and exercise play a significant role in the regulation of arterial function. Systemic acidosis, resulting from respiratory dysfunction (e.g., compromised pulmonary ventilation) or metabolic disorders (e.g., lactic acidosis), induces a moderate elevation of plasma H⁺ concentration. In contrast, localized acidic microenvironments observed in pathological contexts (e.g., ischemic tissues or solid tumors) are primarily driven by compartmentalized proton generation through accelerated glycolytic metabolism (Warburg effect) and dysfunctional ion transport mechanisms. This spatial heterogeneity disrupts physiological pH homeostasis, overwhelming endogenous buffering systems including bicarbonate (HCO3/CO2) and protein-based buffers. In the absence of pathological conditions, exercise is the primary factor responsible for altered pulmonary ventilation and metabolism. This may result in a transient decrease in body pH. The objective of this section is to examine the potential mechanisms through which exercise influences arterial function via proton-sensing GPCRs.

5.1. The Effect of Exercise on the Generation of an Acidic Microenvironment

5.1.1. Regulation of Acid–Base Balance During Respiration and Metabolic Stress

The regulation of the plasma’s acidic environment is primarily maintained by buffer systems (e.g., the bicarbonate buffer system), respiratory control of CO2, and renal excretion of H⁺ and reabsorption of HCO3⁻. At the cellular level, intracellular pH and acid–base balance are regulated by membrane transport proteins, including the sodium–hydrogen antiporter 1 (NHE1), adenosine triphosphatase (ATPase), monocarboxylate transporters (MCT1 and MCT4), as well as Na⁺-HCO3⁻ cotransporters and Cl⁻/HCO3⁻ exchangers [108,109]. Pulmonary ventilation eliminates carbon dioxide (CO2) from venous blood via alveolar gas exchange. Under normal physiological conditions, CO2 produced by muscles during aerobic metabolism is efficiently removed by pulmonary ventilation. However, during lactic acidosis, the buffering of excess H⁺ by bicarbonate (HCO3⁻) generates additional CO2 through the reaction. This increases total CO2 production, potentially overwhelming the compensatory capacity of pulmonary ventilation. Consequently, dissolved CO2 and carbamino-bound CO2 levels rise, further lowering plasma pH [110,111]. Carbonic anhydrase (CA) catalyzes the hydration of CO2 to H⁺ and HCO3⁻, amplifying acidification. While the tricarboxylic acid (TCA) cycle is the primary source of cellular CO2, its overproduction during metabolic stress exacerbates acidosis [112]. During exercise, alterations in the body’s metabolic pattern lead to a decrease in arterial pH due to the limited capacity of pH buffers to regulate and remove the persistently increased levels of H+ in a timely manner [113,114]. Consequently, the regulation of plasma and intracellular acid–base homeostasis is dependent on a variety of mechanisms, including the buffering system, respiratory control, and renal regulation. However, during periods of strenuous exercise or metabolic stress (e.g., lactic acidosis), the overproduction of H+ and CO2 may temporarily exceed the body’s compensatory capacity, resulting in a decrease in arterial pH and an exacerbation of acidosis (see Figure 3).

5.1.2. Exercise and the Regulation of Acid–Base Balance

The duration of the isocapnic buffering period is contingent upon two variables: the quantity of available CO2 and the threshold of arterial carbon dioxide tension (PaCO2) necessary to stimulate ventilation via chemoreceptors [115]. During exercise, CO2 diffuses from the intracellular space into the convective transport medium, namely the blood, which comprises two compartments: plasma and erythrocytes. Subsequently, the CO2 enters the alveoli via the pulmonary capillary barrier and is eliminated from the body via pulmonary gas exchange. Carbon dioxide is transported in physically dissolved, bicarbonate, and carbamate forms [116]. During periods of rest, only 5% of CO2 is stored in its physically dissolved form within the arteries, representing a mere 10% of the total arteriovenous CO2 concentration difference. However, the proportion of CO2 physically dissolved increases to one-third of the total CO2 exchange during exercise [117]. During periods of strenuous exercise, the concentration of carbamates in arterial blood is higher than that in venous blood. Moreover, elevated CO2 partial pressures can precipitate an increase in venous blood carbamates, which can be offset by a reduction in carbamates due to a decline in pH [118]. The concentration of plasma HCO3 levels is 13 times greater than that of physically dissolved CO2. During periods of intense physical exertion, the pH level of the plasma decreases to approximately 7.2, resulting in a 20-fold increase in the concentration of HCO3 [119]. However, the relative contribution of HCO3 to total CO2 exchange is diminished during exercise in comparison to rest. Specifically, HCO3 accounts for only two-thirds of the total CO2 exchange during strenuous exercise, compared to 85% at rest (see Figure 4). Peronnet et al. [120] identified a robust correlation between alterations in arterial blood pH and pulmonary ventilation through empirical investigation. It has been postulated that from the initial ventilation threshold, VT1, to the maximum exercise intensity, the organism regulates plasma acid–base homeostasis by eliminating non-metabolic CO2 from the alkaline reserve.
Furthermore, exercise has been demonstrated to facilitate the accumulation of lactate. It was previously assumed that lactate was the exclusive product of anaerobic metabolism in skeletal muscle. However, recent evidence suggests that it can also be produced during the latter stages of aerobic exercise [121]. The energy molecule adenosine triphosphate (ATP) facilitates muscle contraction by enabling cross-bridge cycling between actin and myosin in the presence of myosin ATPase, which results in muscle stress. However, during sustained exercise, the ATP reserves of skeletal muscle are depleted, necessitating the reliance on phosphocreatine (PCr) and myoglycogen to facilitate the formation of pyruvate and ATP [122]. During strenuous exercise, this pathway is employed to produce ATP, thereby meeting the demands of the cross-bridge cycle and ion pump operation. This necessitates enhanced activity of the Ca2+-ATPase and Na+-K+-ATPase. However, during low-intensity exercise, the mitochondria are unable to fully oxidize the pyruvate produced, resulting in its accumulation in the sarcoplasm and conversion to lactate [123]. Recent studies have demonstrated that lactate can also function as a strong acid anion, promoting the ionization reaction of water and thus the production of H+ [124] (see Figure 4).
A study was conducted in which six male participants engaged in 30 min of resistance and isocaloric endurance exercise. The findings indicate that resistance exercise resulted in elevated lactate levels in comparison to endurance exercise, which in turn led to enhanced metabolic activity during the recovery period [125]. A retrospective analysis revealed that blood lactate concentrations in individuals with multiple sclerosis (MS) were comparable to those of healthy controls during acute sub-extreme and maximal-intensity exercise. However, it was observed that moderate-intensity exercise resulted in a notable reduction in blood lactate levels [126]. Lee et al. [127] conducted experiments demonstrating that both anaerobic and endurance exercise resulted in elevated levels of blood lactate and plasma H+. Furthermore, the administration of short-term creatine has been observed to mitigate the elevation in blood lactate concentrations that occurs in response to late-phase anaerobic or endurance exercise. The results of the Wingate test and incremental tests on cyclists with varying exercise levels and aerobic capacities indicate that both arterial capillary lactate and H+ concentrations were elevated three minutes after exercise. Additionally, mountain bikers demonstrated more pronounced anaerobic effects during incremental tests in comparison to road cyclists [128]. Both the moderate repetition protocol (MRP) and the high repetition protocol (HRP) resulted in an increase in blood lactate concentrations and a decrease in blood pH. The effect was more pronounced with HRP than with MRP. The elevation in blood lactate concentration and the decrease in blood pH were more pronounced with HRP than with MRP [129]. The administration of high-intensity interval training (HIIT) resulted in a notable elevation in blood lactate levels and a considerable augmentation in acidic ion pressure during the exercise period [130]. Prior research has indicated that lactate accumulation is directly linked to the production of H+, which causes a decrease in intramuscular pH or acidosis [131]. In conclusion, the data demonstrate a significant increase in blood lactate levels during both anaerobic and late aerobic exercise, which was accompanied by a subsequent rise in plasma H+ levels, and a corresponding reduction in the body’s circulating pH. The manner in which exercise affects circulating pH through lactate secretion is analogous to the effects of pulmonary ventilation. However, high-intensity exercise may result in a reduction in plasma pH, which is linked to unfavorable conditions such as metabolic acidosis within the body.
During low to moderate-intensity exercise, the body maintains a stable PaCO2 level due to balanced CO2 production and elimination through increased ventilation. In contrast, high-intensity exercise leads to the accumulation of lactic acid due to anaerobic metabolism, resulting in a reduction in plasma pH and the development of metabolic acidosis. The effects of exercise of differing intensities on the accumulation of protons within the organism are not uniform. In general terms, during moderate-intensity exercise, the body’s plasma proton concentration increases to a certain extent, yet it remains within a relatively stable and adjustable range. During this process, the body is able to make full use of oxygen, thus gradually oxidizing and breaking down carbohydrates, fats, and other nutrients. The rate of proton production is relatively smooth and slow, and aerobic metabolism has well-developed metabolic pathways and buffer systems to deal with the small number of protons produced by metabolism [132]. Intracellularly produced protons can be transported and neutralized by proton transport proteins (e.g., NHE1, etc.) in the cell membrane and buffering substances (e.g., HCO3, etc.) in the bloodstream, thereby maintaining relatively stable intracellular and blood proton concentrations [108,109,110]. However, the rate and extent of proton production during high-intensity exercise far exceeds that of moderate-intensity exercise [133]. During periods of high-intensity exercise, there is a significant increase in the incidence of anaerobic metabolism within the body. The process of anaerobic glycolysis produces significant quantities of metabolites, including lactic acid, over a relatively brief period. Concurrently, this process gives rise to substantial H+ release [134,135]. Despite the body’s inherent proton transport and buffering mechanisms, it is unable to effectively respond to the rapid accumulation of large amounts of acidic protons over a short period of time. The intracellular concentration of protons increases rapidly, and there is a tendency for the blood to acidify [136]. In summary, the changes in the body’s proton concentration are relatively moderate during moderate-intensity exercise, whereas proton concentration increases rapidly and substantially during high-intensity exercise, and a longer time is required for subsequent recovery to normal levels.
In addition to exercise, other stressors such as hypoxia, hypertension, and the tumor microenvironment have been demonstrated to affect extracellular proton concentrations. It has been demonstrated that tissue hypoxia can induce glycolysis through the activation of the HIF-1α pathway. This, in turn, results in the production of substantial quantities of lactate and the consequent development of persistent acidosis [137]. In contrast to the phenomenon of exercise-induced acidosis, the condition of hypoxia-associated acidosis is characterized by an imbalance between oxygen supply and demand, accompanied by impaired ATP synthesis and oxidative stress [138]. Furthermore, in hypertension, the vascular endothelium releases protons (via ASIC1 channels) and inflammatory factors under high shear, leading to acidification of the local microenvironment [139,140]. In contrast to the physiological changes that occur in response to exercise, the process of acidification in hypertension represents a pathological alteration, characterized by endothelial dysfunction and vascular remodeling. It has been established that cancer cells within the tumor microenvironment maintain intracellular alkalinization and extracellular acidification by overexpressing CA IX/XII and ATPase, thus promoting invasion and drug resistance [141]. The reverse pH gradient observed within tumors is attributable to malignant metabolic reprogramming. In conclusion, when compared to persistent acidosis triggered by pathological factors such as hypoxia, hypertension, and the tumor microenvironment, the exercise-induced plasma pH change is indicative of enhanced metabolic vitality and physiological stress adaptation of the organism.

5.2. The Potential Mechanisms by Which Exercise Improves Arterial Function Through the Activation of Proton-Sensing GPCRs

The regulation of plasma pH during exercise is contingent upon the equilibrium between CO2 production, irrespective of whether the activity in question is aerobic or anaerobic. The typical range for arterial blood H+ concentrations is 38–40 nmol/L, which corresponds to a pH of approximately 7.42 in a resting state [142]. During exercise, the accumulation of H⁺ in arterial blood primarily results from anaerobic metabolism and lactic acid production. The subsequent increase in ventilation (VE) serves as a compensatory mechanism to mitigate this acidosis by enhancing CO2 excretion, thereby preventing a sustained elevation of arterial H⁺ concentration [143]. The accumulation of plasma H+ following exercise activates proton-sensing GPCRs on cell membranes, particularly VECs and VSMCs. These receptors regulate a number of cellular processes, including proliferation, differentiation, migration, apoptosis, inflammation, and the growth of new blood vessels. Furthermore, they may play a pivotal role in regulating arterial function (see Figure 5).
The extracellular acidic microenvironment has been demonstrated to exert an influence on the activity and function of both VECs and VSMCs [144]. The extant research evidence is exclusively in vitro cellular experimentation. In a study, acid-pretreated ECs were found to prevent autoapoptosis by promoting p38 MAPK and Akt-dependent B-cell lymphoma extra large (Bcl-xL) overexpression [145]. Dong et al. [146] identified that the cAMP/exchange protein activated by 3′-5′-cyclic adenosine monophosphate (Epac) pathway is activated in HUVECs during acidosis, leading to the secretion of VCAM-1 and ICAM-1. The activation of the cAMP/Epac pathway in HUVECs facilitates the secretion of VCAM-1 and ICAM-1. Additionally, it induces the expression of numerous pro-inflammatory genes, including chemokines, cytokines, adhesion molecules, genes associated with the NF-κB pathway, COX-2, and stress-responsive genes. In a study by Namkoong et al. [147], it was demonstrated that forskolin, a cAMP agonist, stimulates angiogenesis by inducing PKA-dependent VEGF expression and Epac-mediated synergistic effects on the downstream PI3K/Akt/endothelial nitric oxide synthase (eNOs) pathway. It has been demonstrated that the acidity of the microenvironment regulates apoptosis, adhesion, and angiogenesis in VECs. Conversely, in VSMCs, an extracellular acidic microenvironment has been observed to increase the expression of MAPK phosphatase 1 (MKP-1), a phosphatase of ERK and p38MAPK, and LPA accumulation in the region of atherosclerotic lesions. This accumulation acted synergistically to promote GPR68-mediated COX-2 and MKP-1 expression [62]. Meanwhile, Tomura et al. [61] discovered that the activation of the OGR1/PLC/Ca2+/extracellular signal-regulated kinase (ERK)/COX/ PGI2 signaling pathway, through the use of siRNAs and specific inhibitor intervention methods, results in an increase in cAMP accumulation and PGI2 production, while simultaneously inhibiting the development of atherogenesis. Furthermore, acidosis has been demonstrated to induce vasodilation by activating eNOS and ATP-sensing potassium channels in the systemic vasculature. This, in turn, results in the hyperpolarization of VSMCs [40,148]. The acidic microenvironment in plasma exerts a considerable influence on vascular inflammation, diastole, and generation by modulating intracellular downstream signaling pathways in VECs and VSMCs (see Figure 5).

6. Conclusions

One of the principal factors responsible for fluctuations in the pH of an organism’s microenvironment is physical exercise. During exercise, alterations in the concentration of carbon dioxide and lactate may result in an increase in the concentration of hydrogen ions in the plasma. Research has indicated that proton-sensing GPCRs expressed on VECs and VSMCs have a significant impact on various arterial functions, including the proliferation, differentiation, migration, apoptosis, and angiogenesis of VECs and VSMCs. Furthermore, these receptors play a role in regulating the expression of inflammatory factors associated with atherosclerotic inflammation, including NF-κB, VCAM-1, ICAM-1, and TNF-α. The majority of previous research into proton-sensing GPCRs has predominantly concentrated on tumors and the perception of alterations in intracellular pH in tumor cells. This review is the inaugural investigation into the regulatory function of activated proton-sensing GPCRs in arterial function and the mechanisms by which they operate within the context of an exercise-mediated plasma acidic microenvironment.
The regulation of arterial function by proton-sensing GPCRs represents a developing area of research that requires further investigation to address certain outstanding issues. Despite the well-established regulation of arterial function by proton-sensing GPCRs, the precise direction and molecular mechanisms regulated by different receptors at varying pH levels remain unclear. This encompasses the specific effects of diverse signaling pathways and the cumulative influence of multiple receptors. The regulatory mechanisms of proton-sensing GPCRs by different intensities of exercise have yet to be elucidated. Nevertheless, the evidence indicates that low- and medium-intensity exercise has a beneficial effect on arterial function, whereas high-intensity resistance exercise may result in augmented arterial stiffness. The impact of low circulating pH on arterial function following high-intensity exercise remains unclear. The currently available evidence pertains to the regulation of plasma pH by exercise and the influence of plasma pH on arterial function via proton-sensing GPCRs. Despite the existence of stratified studies, there is currently no experimental investigation that has evaluated whether exercise affects arterial function through direct modification of plasma pH via proton-sensing GPCRs.
The emerging role of proton-sensing GPCRs as pH-dependent regulators of arterial homeostasis establishes a novel paradigm for understanding exercise-mediated cardiovascular adaptation. Further research is required to elucidate the precise mechanism through which the exercise microenvironment regulates arterial function by activating proton-sensing GPCRs. This would provide a scientific basis for understanding the role of exercise in preventing arterial dysfunction at the molecular level. In turn, this would provide a reliable foundation for the development of medications and exercise prescriptions to treat arterial stiffness. Furthermore, disparities have been observed in the efficiency of the intracellular acid buffering system and proton transport mechanisms across various age groups. Intrinsic health variations and genetic distinctions have also been demonstrated to influence the number and activity of intracellular proton transport proteins, as well as the structure and function of proton-sensing GPCRs. From a sports medicine perspective, pharmaceuticals developed to regulate the activity of proton-sensing GPCRs in patients with arterial dysfunction may have different effects in patients with different genetic backgrounds, health levels, and ages. Consequently, the potential for disparities in exercise-induced alterations in proton concentration and proton-sensing GPCR activity based on individual variation in genetics, health levels, and ages merits further investigation.
Table 1. Signaling and pharmacological reagents of proton-sensing GPCRs.
Table 1. Signaling and pharmacological reagents of proton-sensing GPCRs.
ReceptorLocationSignalingProtonsAgonist (EC50)Antagonist (IC50)Ref.
GPR68 (OGR1)Human:14q32.11 NM_003485
Mouse: Chr 12 NM_175493
Gαq;
Gαs;
Gαi;
G12/13
pH 7.8–5.6, maximum activity at pH 6.8CARTPT (3.2 µM) MOsteocrin (0.4 µM); MCorticotropin (1.8 µM); 3,5-disubstituted isoxazoles (µM range); CART (1 µM); Pro-opiomelanocortin-derived peptide (1.3 µM)Cu2+ (µM range); Zn2+ (µM range)[44,149,150]
GPR4Human: 19q13.32 NM_005282
Mouse: Chr 7 NM_175668
Gαs;
Gαq;
Gαi;
G12/13
pH 7.6–5.6NDCompound 3b (67 nM); NE 52-QQ 57 (70 nM); NE 52-QQ57 (70 nM); Compound 39c (110 nM)[44,141,142,143,144,145,146,147,148,149,150,151,152,153,154]
GPR132
(G2A)
Human:14q32.33 NM_001278695.2
Mouse: Chr 12 NM_019925
Gαs;
Gαq
pH 8.2–6.69S-HODE (~0.5 µM); 11-HETE (~1 µM); N -palmitoylglycine (~800 nM); N-linoleoylglycine (~800 nM); ONC212 (~400 nM); 11,12-EET (~10 µM); 9,10-EpOME (~10 µM)Lysophosphatidylcholine (~10 µM); Telmisartan (~10 µM); GSK1820795A (~1 µM)[155,156,157,158,159]
GPR65 (TDAG8)Human: 14q31.3 NM_003608
Mouse: Chr 12 NM_ 008152.3
GαspH 7.2–5.7Psychosine (3.4 µM); BTB09089 (active concentration > 5 µM)ND[81,160,161,162]
Note: ND, not determined.
Table 2. Regulation of arterial function by exercise.
Table 2. Regulation of arterial function by exercise.
SubjectsExercise Intervention ProgramMode of ActionRef.
ModelCharacteristicsSample SizeTypeIntensityDuration
C57BL/6, WT mice; ApoEtm1Unc, ApoE KO mice5–6 weeks oldNDTreadmill running15 m/min at a 5° grade (60–80% of VO2max)5 days/week; 60 min/session; 15–16 weeksEndothelial dysfunction ↑[103]
Patients with amnestic MCI 29 in SAT/19 in AET70Moderate-to-vigorous AET or stretching and toning (SAT)Moderate to vigorous12 monthsVO2peak ↑;
carotid β-stiffness index ↑;
CBF pulsatility ↑
[105]
Older sedentary overweight and obese men67 ± 2 years, BMI: 30.3 ± 2.8 kg/m217 malesProgressive, aerobic exercise70% maximal power3 days/week; 50 min/session; 8 weeksEndothelial function ↑, retinal arteriolar width ↑, cardiovascular risk ↓[163]
Recreational athletes45.9 ± 9.6 years46 females/5 malesEndurance exerciseNDNDCoronary artery calcification →[164]
Young menTealthy and recreationally active; 23 ± 2 years)10 malesIncremental leg cycling exercise50, 100, 150 Watts30 minRadial artery mean and anterograde SR ↑[165]
Overweight men21–30 years; BMI: 30 ± 3 kg/m28 malesSwimming training50–80% HRmax3 days/week; 55 min/session; 8 weeksCarotid arterial stiffness, systolic blood pressure, Peripheral resistance ↓; blood flow velocity, flow rate, maximal, mean wall shear stress ↑[166]
Healthy adults22 ± 2 years; BMI: 22 ± 2 kg/m212Cycling85 ± 5% HRmax30 minICA conductance ↑; vasodilation of the ICA ↑[167]
Healthy adults23 ± 4 years11 females/9 malesIsometric handgrip training30% of maximal voluntary contraction3 days/week; four, 2 min unilateral contractions; 8 weeksEndothelium-dependent vasodilation ↑[168]
Healthy adults61.0 ± 1.3 years60Aerobic exerciseMedium–high intensity8 weeksArterial stiffness ↓[169]
Overweight and obese adultsBMI: 30.5 ± 7.230Aerobic exerciseND8 weeksArterial dysfunction ↓[170]
Healthy adults61 ± 2 years4 females/7 malesEndurance exercise70% VO2max60 min/session; 10 daysFMD ↑; CAC ↑[171]
Healthy adults66 ± 1 years5 females/6 malesRecumbent cycling75–80% HRmax30 minBrachial artery FMD ↑[172]
Healthy adultsYA: 26 ± 5 years; 23.8 ± 3.3 kg/m2
OA: 60 ± 6 years; 30.0 ± 5.5 kg/m2
21 young adults; 25 older adultsUnilateral maximal isokinetic knee flexion/extension exercise1RM3 sets; 10 repsCCA strain time ↓[173]
Patients with metabolic syndrome 51 ± 12 years57Endurance exercise60–85% 1RM8 weekscfPWV ↓; artery stiffness →[107]
Healthy adults18–30 years14 females/12 malesResistance exercise75% 1RM3 sets; 10 repsArterial stiffness↑[174]
Healthy adults24 ± 1 year7 malesEccentric exerciseHigh intensity1 set; 50 repsCarotid arterial compliance ↓; endothelial function ↓; β-stiffness index ↑[175]
Sprague–Dawley rats10 weeks old40 malesTreadmill running;
HIIT
30 m/min; High intensity5 days/week, 60 min/ session; 8 weeks; 4 days/week, 8 weeks, 14 repeats of 20 s/session, 10 s pause between sessionsPWV ↑; central arterial stiffness ↓[176]
Patients with CAD 71.8 ± 10.2 years18Endurance training;
HIIT
60%; 85–90% HRmax30 min; 10 interval training periodsAcute endurance training; AS ↓; resistance training AS ↑[177]
Healthy adults56 ± 5 years25 femalesEndurance exercise; resistance exerciseMedium to high strength150 min/weeks endurance exercise; 2 or more days/weeks strength-based exercisePCS ↑; PSR ↑[178]
Patients with peripheral artery disease 50–80 years12Walking exercise; resistance exercise; combined exerciseND10 bouts of 2 min walking; 2 sets of 10 reps in 8 resistance exercises; 1 set of 10 reps in 8 resistance exercises + 5 bouts of 2 min walkingArtery stiffness ↓[179]
Same-sex twins31 monozygotic, 14 dizygotic pairs; 25.8 ± 6.0 years90Endurance exercise; resistance exerciseND3 monthsFMD ↑; vascular function ↑[180]
Patients with chronic kidney disease 55 years and older; CKD stages 3b–499Endurance exercise; resistance exercise40–70% HRmax6 days/week, 90 min/session; 12 monthsArterial function ↑[106]
Patients with bariatric surgery 8–45 years40 femalesEndurance exercise; resistance exerciseModerate intensity; 50–75% 1RM3 days/week; 60 min/session; 16 monthsArterial stiffness ↓[181]
Sedentary older adults64 ± 1 years64MICT; HIIT70% HRmax; 4 × 4 min at 90% HRmax interspersed with 3 × 3 min active recovery at 70% HRmax4 days/week; 8 weeksMICT: carotid artery compliance ↑; cfPWV ↑
HIIT: carotid artery compliance →; cfPWV →
[182]
Healthy adults23.5 ± 1.2 years10MICT; HIIT40% HRmax; 85% HRmaxMICT: 40 min;
HIIT: 1 min/session; 2 min between sets; total 26 min
Artery blood flow velocity →[183]
Healthy adults21.4 ± 0.8 years;
1.73 ± 0.03 m; 62.1 ± 6.4 kg
11 malesInterval training; interval exercise of semi-recumbent cycling57.6 kJ/exercise session12 minICA SR↑[184]
Note: “↑”: increase; “↓”: decrease; “→”: no significant change; HRmax: maximal heart rate; VO2max: maximal oxygen uptake; 1RM: maximal force for 1 repetition; ND, not determined; VO2peak: peak oxygen uptake; CBF: cerebral blood flow; ICA: internal carotid artery; SR: shear rate; L-FMC: low-flow mediated constriction; FMD: low mediated dilatation; PWV: pulse wave velocity; BMI: body mass index; CAD: coronary artery disease; AS: arterial stiffness; HIIT: high-intensity interval training; MICT: moderate-intensity continuous training; PWV: pulse wave velocity; FMD: flow-mediated dilation; CAC: circulating angiogenic cell; CCA: common carotid artery; PCS: peak circumferential strain; PSR: peak strain rate.

Funding

This work was supported in part by a grant from the National Natural Science Foundation of China (32271226), the National Key R&D Program of China (2020YFA0803800), the Key Laboratory of Exercise and Health Sciences (Shanghai University of Sport), Ministry of Education, and the Shanghai Baiyulan Talent Program Pujiang Project Funding (ID: 24PJC067).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

ACadenylate cyclase
AMPKAMP-activated protein kinase
AoSMCsaortic smooth muscle cells
ASICsacid-sensitive ion channels
ATF3activating transcription factor 3
ATPadenosine triphosphate
ATPaseadenosine triphosphatase
Bcl-xLB-cell lymphoma extra large
CAcarbonic anhydrase
Calmcalmodulin
cAMPcyclic adenosine monophosphate
CBXcarotid body excision
CHOPC/EBP homologous protein
CO2carbon dioxide
COXcyclooxygenase
CT1-α1collagen type 1-α1
CVDscardiovascular diseases
DAGdiacylglycerol
DDIT3DNA damage-induced transcript 3
eNOsendothelial nitric oxide synthase
Epacexchange protein activated by 3′–5′-cyclic adenosine monophosphate
EPCsendothelial progenitor cells
ERKextracellular signal-regulated kinase
FICO2fraction of inspired CO2
G2AG2 accumulation protein
GRKG protein-coupled receptor kinase
GTPguanosine triphosphate
GTPaseguanosine triphosphatase
H+hydrion
H2Owater molecule
HbNH2aminohemoglobin
HbNHCOOHcarbamino hemoglobin
HbO2oxyhemoglobin
HCO3bicarbonate radical
HDLhigh-density lipoprotein
HHbunionized hemoglobin
HIIThigh-intensity interval training
HRPhigh repetition protocol
HUVECshuman umbilical vein endothelial cells
ICAM-1intercellular adhesion molecule-1
IL-6interleukin-6
IP3inositol triphosphate
KHCO3potassium bicarbonate
KIM-1kidney injury molecule 1
KLF2Kruppel-like factor 2
KOknockout
MAPKmitogen-activated protein kinase
MCAvmiddle cerebral artery blood velocity
MCT1monocarboxylate transporter
MImyocardial infarction
MKP-1MAPK phosphatase 1
MRPmoderate repetition protocol
MSmultiple sclerosis
Na+/K+-ATPaseSodium–potassium ATPase
NAD+nicotinamide adenine dinucleotide
NADHreduced form of nicotinamide–adenine dinucleotide
NaHCO3sodium hydrogen carbonate
NFKB1nuclear factor kappa B subunit 1
NF-κBnuclear factor k-binding
NHE1sodium–hydrogen antiporter 1
NLRP3NOD-, LRR-, and pyrin domain-containing protein 3
Notch1notch receptor 1
OGR1ovarian cancer G protein-coupled receptor
OSSoscillatory shear stress
PaCO2carbon dioxide tension
PCNAproliferating cell nuclear antigen
PCrphosphocreatine
PDKphosphoinositide-dependent protein kinase
PGI2prostaglandin-I-2
PI3Kphosphatidylinositol 3-hydroxy kinase
PIP2guanosine triphosphatase
PKAprotein kinase A
PKCprotein kinase C
PLCphospholipase C
proton-sensing GPCRsproton-sensing G protein-coupled receptors
RasRat sarcoma
RCrespiratory compensation threshold
RELBRELB proto-oncogene, NF-KB subunit
RhoRas homologous
ROCKRho-associated protein kinase
sGCsoluble guanylyl cyclase
STAT3signal transducer and activator of transcription 3
TDAG8T-cell death-associated gene 8
TNF-αtumor necrosis factor-α
VCAM-1vascular cell adhesion molecule-1
VEpulmonary ventilation
VECsvascular endothelial cells
VEGFAvascular endothelial growth factor A
VSMCsvascular smooth muscle cells
7TMRs7-transmembrane structural domain receptors

References

  1. Townsend, N.; Kazakiewicz, D.; Lucy Wright, F.; Timmis, A.; Huculeci, R.; Torbica, A.; Gale, C.P.; Achenbach, S.; Weidinger, F.; Vardas, P. Epidemiology of cardiovascular disease in Europe. Nat. Rev. Cardiol. 2022, 19, 133–143. [Google Scholar] [CrossRef] [PubMed]
  2. Zhang, Z.; Zou, Y.; Song, C.; Cao, K.; Cai, K.; Chen, S.; Wu, Y.; Geng, D.; Sun, G.; Zhang, N.; et al. Advances in the study of exosomes in cardiovascular diseases. J. Adv. Res. 2024, 66, 133–153. [Google Scholar] [CrossRef]
  3. Chong, B.; Jayabaskaran, J.; Jauhari, S.M.; Chan, S.P.; Goh, R.; Kueh, M.T.W.; Li, H.Y.; Chin, Y.H.; Kong, G.; Anand, V.V.; et al. Global burden of cardiovascular diseases: Projections from 2025 to 2050. Eur. J. Prev. Cardiol. 2024, 13, zwae281. [Google Scholar] [CrossRef]
  4. Boutouyrie, P.; Chowienczyk, P.; Humphrey, J.D.; Mitchell, G.F. Arterial Stiffness and Cardiovascular Risk in Hypertension. Circ. Res. 2021, 128, 864–886. [Google Scholar] [CrossRef] [PubMed]
  5. Motau, T.H.; Norton, G.R.; Sareli, P.; Woodiwiss, A.J. Aortic Pulse Pressure Does Not Adequately Index Cardiovascular Risk Factor-Related Changes in Aortic Stiffness and Forward Wave Pressure. Am. J. Hypertens. 2018, 31, 981–987. [Google Scholar] [CrossRef] [PubMed]
  6. Gimbrone, M.A., Jr.; Garcia-Cardena, G. Endothelial Cell Dysfunction and the Pathobiology of Atherosclerosis. Circ. Res. 2016, 118, 620–636. [Google Scholar] [CrossRef]
  7. Xu, S.; Ilyas, I.; Little, P.J.; Li, H.; Kamato, D.; Zheng, X.; Luo, S.; Li, Z.; Liu, P.; Han, J.; et al. Endothelial Dysfunction in Atherosclerotic Cardiovascular Diseases and Beyond: From Mechanism to Pharmacotherapies. Pharmacol. Rev. 2021, 73, 924–967. [Google Scholar] [CrossRef]
  8. Perry, A.S.; Dooley, E.E.; Master, H.; Spartano, N.L.; Brittain, E.L.; Pettee Gabriel, K. Physical Activity Over the Lifecourse and Cardiovascular Disease. Circ. Res. 2023, 132, 1725–1740. [Google Scholar] [CrossRef]
  9. Valenzuela, P.L.; Ruilope, L.M.; Santos-Lozano, A.; Wilhelm, M.; Krankel, N.; Fiuza-Luces, C.; Lucia, A. Exercise benefits in cardiovascular diseases: From mechanisms to clinical implementation. Eur. Heart J. 2023, 44, 1874–1889. [Google Scholar] [CrossRef]
  10. Howangyin, K.Y.; Silvestre, J.S. Diabetes mellitus and ischemic diseases: Molecular mechanisms of vascular repair dysfunction. Arterioscler. Thromb. Vasc. Biol. 2014, 34, 1126–1135. [Google Scholar] [CrossRef]
  11. Khalafi, M.; Symonds, M.E.; Sakhaei, M.H.; Ghasemi, F. The effects of exercise training on circulating adhesion molecules in adults: A systematic review and meta-analysis. PLoS ONE 2023, 18, e0292734. [Google Scholar] [CrossRef]
  12. Yang, A.L.; Chen, H.I. Chronic exercise reduces adhesion molecules/iNOS expression and partially reverses vascular responsiveness in hypercholesterolemic rabbit aortae. Atherosclerosis 2003, 169, 11–17. [Google Scholar] [CrossRef] [PubMed]
  13. Adams, V.; Linke, A.; Krankel, N.; Erbs, S.; Gielen, S.; Mobius-Winkler, S.; Gummert, J.F.; Mohr, F.W.; Schuler, G.; Hambrecht, R. Impact of regular physical activity on the NAD(P)H oxidase and angiotensin receptor system in patients with coronary artery disease. Circulation 2005, 111, 555–562. [Google Scholar] [CrossRef]
  14. Konigstein, K.; Dipla, K.; Zafeiridis, A. Training the Vessels: Molecular and Clinical Effects of Exercise on Vascular Health-A Narrative Review. Cells 2023, 12, 2544. [Google Scholar] [CrossRef]
  15. Conraads, V.M.; Beckers, P.; Bosmans, J.; De Clerck, L.S.; Stevens, W.J.; Vrints, C.J.; Brutsaert, D.L. Combined endurance/resistance training reduces plasma TNF-alpha receptor levels in patients with chronic heart failure and coronary artery disease. Eur. Heart J. 2002, 23, 1854–1860. [Google Scholar] [CrossRef] [PubMed]
  16. Munk, P.S.; Breland, U.M.; Aukrust, P.; Ueland, T.; Kvaloy, J.T.; Larsen, A.I. High intensity interval training reduces systemic inflammation in post-PCI patients. Eur. J. Cardiovasc. Prev. Rehabil. 2011, 18, 850–857. [Google Scholar] [CrossRef]
  17. Yoshikawa, D.; Ishii, H.; Kurebayashi, N.; Sato, B.; Hayakawa, S.; Ando, H.; Hayashi, M.; Isobe, S.; Okumura, T.; Hirashiki, A.; et al. Association of cardiorespiratory fitness with characteristics of coronary plaque: Assessment using integrated backscatter intravascular ultrasound and optical coherence tomography. Int. J. Cardiol. 2013, 162, 123–128. [Google Scholar] [CrossRef] [PubMed]
  18. Madssen, E.; Moholdt, T.; Videm, V.; Wisloff, U.; Hegbom, K.; Wiseth, R. Coronary atheroma regression and plaque characteristics assessed by grayscale and radiofrequency intravascular ultrasound after aerobic exercise. Am. J. Cardiol. 2014, 114, 1504–1511. [Google Scholar] [CrossRef]
  19. Haskell, W.L.; Sims, C.; Myll, J.; Bortz, W.M.; St Goar, F.G.; Alderman, E.L. Coronary artery size and dilating capacity in ultradistance runners. Circulation 1993, 87, 1076–1082. [Google Scholar] [CrossRef]
  20. Nguyen, P.K.; Terashima, M.; Fair, J.M.; Varady, A.; Taylor-Piliae, R.E.; Iribarren, C.; Go, A.S.; Haskell, W.L.; Hlatky, M.A.; Fortmann, S.P.; et al. Physical activity in older subjects is associated with increased coronary vasodilation: The ADVANCE study. JACC Cardiovasc. Imaging 2011, 4, 622–629. [Google Scholar] [CrossRef]
  21. Green, D.J.; Hopman, M.T.; Padilla, J.; Laughlin, M.H.; Thijssen, D.H. Vascular Adaptation to Exercise in Humans: Role of Hemodynamic Stimuli. Physiol. Rev. 2017, 97, 495–528. [Google Scholar] [CrossRef] [PubMed]
  22. Thijssen, D.H.; Cable, N.T.; Green, D.J. Impact of exercise training on arterial wall thickness in humans. Clin. Sci. (Lond.) 2012, 122, 311–322. [Google Scholar] [CrossRef] [PubMed]
  23. Constantin-Teodosiu, D.; Cederblad, G.; Bergstrom, M.; Greenhaff, P.L. Maximal-intensity exercise does not fully restore muscle pyruvate dehydrogenase complex activation after 3 days of high-fat dietary intake. Clin. Nutr. 2019, 38, 948–953. [Google Scholar] [CrossRef]
  24. Terpe, P.; Ruhs, S.; Dubourg, V.; Gekle, M.; Bucher, M. The synergism of cytosolic acidosis and reduced NAD/NADH ratio is responsible for lactic acidosis-induced vascular smooth muscle cell impairment in sepsis. J. Biomed. Sci. 2024, 31, 3. [Google Scholar] [CrossRef] [PubMed]
  25. Xu, J.; Mathur, J.; Vessieres, E.; Hammack, S.; Nonomura, K.; Favre, J.; Grimaud, L.; Petrus, M.; Francisco, A.; Li, J.; et al. GPR68 Senses Flow and Is Essential for Vascular Physiology. Cell 2018, 173, 762–775.e716. [Google Scholar] [CrossRef]
  26. Krewson, E.A.; Sanderlin, E.J.; Marie, M.A.; Akhtar, S.N.; Velcicky, J.; Loetscher, P.; Yang, L.V. The Proton-Sensing GPR4 Receptor Regulates Paracellular Gap Formation and Permeability of Vascular Endothelial Cells. iScience 2020, 23, 100848. [Google Scholar] [CrossRef]
  27. Bolick, D.T.; Skaflen, M.D.; Johnson, L.E.; Kwon, S.C.; Howatt, D.; Daugherty, A.; Ravichandran, K.S.; Hedrick, C.C. G2A deficiency in mice promotes macrophage activation and atherosclerosis. Circ. Res. 2009, 104, 318–327. [Google Scholar] [CrossRef]
  28. Chen, L.D.; Zhu, W.T.; Cheng, Y.Y.; Li, Z.H.; Chen, Y.Q.; Yuan, Z.W.; Lin, C.Y.; Jing, D.D.; Liu, Z.Q.; Yan, P.K. T-cell death-associated gene 8 accelerates atherosclerosis by promoting vascular smooth muscle cell proliferation and migration. Atherosclerosis 2020, 297, 64–73. [Google Scholar] [CrossRef]
  29. Yang, K.; Fan, M.; Wang, X.; Xu, J.; Wang, Y.; Gill, P.S.; Ha, T.; Liu, L.; Hall, J.V.; Williams, D.L.; et al. Lactate induces vascular permeability via disruption of VE-cadherin in endothelial cells during sepsis. Sci. Adv. 2022, 8, eabm8965. [Google Scholar] [CrossRef]
  30. Imenez Silva, P.H.; Wagner, C.A. Physiological relevance of proton-activated GPCRs. Pflugers Arch. 2022, 474, 487–504. [Google Scholar] [CrossRef]
  31. von Breitenbuch, P.; Kurz, B.; Wallner, S.; Zeman, F.; Brochhausen, C.; Schlitt, H.J.; Schreml, S. Expression of pH-Sensitive GPCRs in Peritoneal Carcinomatosis of Colorectal Cancer-First Results. J. Clin. Med. 2023, 12, 1803. [Google Scholar] [CrossRef] [PubMed]
  32. Howard, M.K.; Hoppe, N.; Huang, X.P.; Macdonald, C.B.; Mehrota, E.; Grimes, P.R.; Zahm, A.; Trinidad, D.D.; English, J.; Coyote-Maestas, W.; et al. Molecular basis of proton-sensing by G protein-coupled receptors. bioRxiv 2024. [Google Scholar] [CrossRef]
  33. Hopkins, P.N. Molecular biology of atherosclerosis. Physiol. Rev. 2013, 93, 1317–1542. [Google Scholar] [CrossRef]
  34. Mohanty, I.; Parija, S.C.; Suklabaidya, S.; Rattan, S. Acidosis potentiates endothelium-dependent vasorelaxation and gap junction communication in the superior mesenteric artery. Eur. J. Pharmacol. 2018, 827, 22–31. [Google Scholar] [CrossRef]
  35. Grogan, A.; Lucero, E.Y.; Jiang, H.; Rockman, H.A. Pathophysiology and pharmacology of G protein-coupled receptors in the heart. Cardiovasc. Res. 2023, 119, 1117–1129. [Google Scholar] [CrossRef]
  36. Casimir, G.J.; Lefevre, N.; Corazza, F.; Duchateau, J.; Chamekh, M. The Acid-Base Balance and Gender in Inflammation: A Mini-Review. Front. Immunol. 2018, 9, 475. [Google Scholar] [CrossRef]
  37. de Valliere, C.; Wang, Y.; Eloranta, J.J.; Vidal, S.; Clay, I.; Spalinger, M.R.; Tcymbarevich, I.; Terhalle, A.; Ludwig, M.G.; Suply, T.; et al. G Protein-coupled pH-sensing Receptor OGR1 Is a Regulator of Intestinal Inflammation. Inflamm. Bowel Dis. 2015, 21, 1269–1281. [Google Scholar] [CrossRef] [PubMed]
  38. Nassios, A.; Wallner, S.; Haferkamp, S.; Klingelhoffer, C.; Brochhausen, C.; Schreml, S. Expression of proton-sensing G-protein-coupled receptors in selected skin tumors. Exp. Dermatol. 2019, 28, 66–71. [Google Scholar] [CrossRef] [PubMed]
  39. Rowe, J.B.; Kapolka, N.J.; Taghon, G.J.; Morgan, W.M.; Isom, D.G. The evolution and mechanism of GPCR proton sensing. J. Biol. Chem. 2021, 296, 100167. [Google Scholar] [CrossRef]
  40. Okajima, F. Regulation of inflammation by extracellular acidification and proton-sensing GPCRs. Cell Signal 2013, 25, 2263–2271. [Google Scholar] [CrossRef]
  41. Sisignano, M.; Fischer, M.J.M.; Geisslinger, G. Proton-Sensing GPCRs in Health and Disease. Cells 2021, 10, 2050. [Google Scholar] [CrossRef] [PubMed]
  42. Jiang, H.; Galtes, D.; Wang, J.; Rockman, H.A. G protein-coupled receptor signaling: Transducers and effectors. Am. J. Physiol. Cell Physiol. 2022, 323, C731–C748. [Google Scholar] [CrossRef] [PubMed]
  43. Imenez Silva, P.H.; Camara, N.O.; Wagner, C.A. Role of proton-activated G protein-coupled receptors in pathophysiology. Am. J. Physiol. Cell Physiol. 2022, 323, C400–C414. [Google Scholar] [CrossRef] [PubMed]
  44. Ludwig, M.G.; Vanek, M.; Guerini, D.; Gasser, J.A.; Jones, C.E.; Junker, U.; Hofstetter, H.; Wolf, R.M.; Seuwen, K. Proton-sensing G-protein-coupled receptors. Nature 2003, 425, 93–98. [Google Scholar] [CrossRef]
  45. Morales Rodriguez, L.M.; Crilly, S.E.; Rowe, J.B.; Isom, D.G.; Puthenveedu, M.A. Location-biased activation of the proton-sensor GPR65 is uncoupled from receptor trafficking. Proc. Natl. Acad. Sci. USA 2023, 120, e2302823120. [Google Scholar] [CrossRef]
  46. Huang, X.P.; Kenakin, T.P.; Gu, S.; Shoichet, B.K.; Roth, B.L. Differential Roles of Extracellular Histidine Residues of GPR68 for Proton-Sensing and Allosteric Modulation by Divalent Metal Ions. Biochemistry 2020, 59, 3594–3614. [Google Scholar] [CrossRef]
  47. Ren, J.; Zhang, L. Effects of ovarian cancer G protein coupled receptor 1 on the proliferation, migration, and adhesion of human ovarian cancer cells. Chin. Med. J. 2011, 124, 1327–1332. [Google Scholar]
  48. Tobo, M.; Tomura, H.; Mogi, C.; Wang, J.Q.; Liu, J.P.; Komachi, M.; Damirin, A.; Kimura, T.; Murata, N.; Kurose, H.; et al. Previously postulated “ligand-independent” signaling of GPR4 is mediated through proton-sensing mechanisms. Cell Signal 2007, 19, 1745–1753. [Google Scholar] [CrossRef]
  49. Xiao, R.; Liu, J.; Shawn Xu, X.Z. Mechanosensitive GPCRs and ion channels in shear stress sensing. Curr. Opin. Cell Biol. 2023, 84, 102216. [Google Scholar] [CrossRef]
  50. Weiss, K.T.; Fante, M.; Kohl, G.; Schreml, J.; Haubner, F.; Kreutz, M.; Haverkampf, S.; Berneburg, M.; Schreml, S. Proton-sensing G protein-coupled receptors as regulators of cell proliferation and migration during tumor growth and wound healing. Exp. Dermatol. 2017, 26, 127–132. [Google Scholar] [CrossRef]
  51. Chen, X.; Jaiswal, A.; Costliow, Z.; Herbst, P.; Creasey, E.A.; Oshiro-Rapley, N.; Daly, M.J.; Carey, K.L.; Graham, D.B.; Xavier, R.J. pH sensing controls tissue inflammation by modulating cellular metabolism and endo-lysosomal function of immune cells. Nat. Immunol. 2022, 23, 1063–1075. [Google Scholar] [CrossRef] [PubMed]
  52. Tcymbarevich, I.V.; Eloranta, J.J.; Rossel, J.B.; Obialo, N.; Spalinger, M.; Cosin-Roger, J.; Lang, S.; Kullak-Ublick, G.A.; Wagner, C.A.; Scharl, M.; et al. The impact of the rs8005161 polymorphism on G protein-coupled receptor GPR65 (TDAG8) pH-associated activation in intestinal inflammation. BMC Gastroenterol. 2019, 19, 2. [Google Scholar] [CrossRef]
  53. Liu, J.P.; Nakakura, T.; Tomura, H.; Tobo, M.; Mogi, C.; Wang, J.Q.; He, X.D.; Takano, M.; Damirin, A.; Komachi, M.; et al. Each one of certain histidine residues in G-protein-coupled receptor GPR4 is critical for extracellular proton-induced stimulation of multiple G-protein-signaling pathways. Pharmacol. Res. 2010, 61, 499–505. [Google Scholar] [CrossRef] [PubMed]
  54. Li, Q.; Feng, C.; Li, L.; Xu, G.; Gu, H.; Li, S.; Li, D.; Liu, M.; Han, S.; Zheng, B. Lipid Receptor G2A-Mediated Signal Pathway Plays a Critical Role in Inflammatory Response by Promoting Classical Macrophage Activation. J. Immunol. 2021, 206, 2338–2352. [Google Scholar] [CrossRef]
  55. de Valliere, C.; Cosin-Roger, J.; Simmen, S.; Atrott, K.; Melhem, H.; Zeitz, J.; Madanchi, M.; Tcymbarevich, I.; Fried, M.; Kullak-Ublick, G.A.; et al. Hypoxia Positively Regulates the Expression of pH-Sensing G-Protein-Coupled Receptor OGR1 (GPR68). Cell Mol. Gastroenterol. Hepatol. 2016, 2, 796–810. [Google Scholar] [CrossRef]
  56. Assimos, D.G. Re: The Proton-Activated Ovarian Cancer G Protein-Coupled Receptor 1 (OGR1) is Responsible for Renal Calcium Loss during Acidosis. J. Urology 2020, 204, 380–381. [Google Scholar] [CrossRef] [PubMed]
  57. Krieger, N.S.; Chen, L.; Becker, J.; Chan, M.; Bushinsky, D.A. Effect of Osteoblast-Specific Deletion of the Proton Receptor OGR1. JBMR Plus 2022, 6, e10691. [Google Scholar] [CrossRef]
  58. de Valliere, C.; Cosin-Roger, J.; Baebler, K.; Schoepflin, A.; Mamie, C.; Mollet, M.; Schuler, C.; Bengs, S.; Lang, S.; Scharl, M.; et al. pH-Sensing G Protein-Coupled Receptor OGR1 (GPR68) Expression and Activation Increases in Intestinal Inflammation and Fibrosis. Int. J. Mol. Sci. 2022, 23, 1419. [Google Scholar] [CrossRef]
  59. Kadowaki, M.; Yamada, H.; Sato, K.; Shigemi, H.; Umeda, Y.; Morikawa, M.; Waseda, Y.; Anzai, M.; Kamide, Y.; Aoki-Saito, H.; et al. Extracellular acidification-induced CXCL8 production through a proton-sensing receptor OGR1 in human airway smooth muscle cells: A response inhibited by dexamethasone. J. Inflamm. 2019, 16, 4. [Google Scholar] [CrossRef]
  60. de Valliere, C.; Vidal, S.; Clay, I.; Jurisic, G.; Tcymbarevich, I.; Lang, S.; Ludwig, M.G.; Okoniewski, M.; Eloranta, J.J.; Kullak-Ublick, G.A.; et al. The pH-sensing receptor OGR1 improves barrier function of epithelial cells and inhibits migration in an acidic environment. Am. J. Physiol. Gastrointest. Liver Physiol. 2015, 309, G475–G490. [Google Scholar] [CrossRef] [PubMed]
  61. Tomura, H.; Wang, J.Q.; Komachi, M.; Damirin, A.; Mogi, C.; Tobo, M.; Kon, J.; Misawa, N.; Sato, K.; Okajima, F. Prostaglandin I(2) production and cAMP accumulation in response to acidic extracellular pH through OGR1 in human aortic smooth muscle cells. J. Biol. Chem. 2005, 280, 34458–34464. [Google Scholar] [CrossRef]
  62. Liu, J.P.; Komachi, M.; Tomura, H.; Mogi, C.; Damirin, A.; Tobo, M.; Takano, M.; Nochi, H.; Tamoto, K.; Sato, K.; et al. Ovarian cancer G protein-coupled receptor 1-dependent and -independent vascular actions to acidic pH in human aortic smooth muscle cells. Am. J. Physiol. Heart Circ. Physiol. 2010, 299, H731–H742. [Google Scholar] [CrossRef]
  63. Ding, S.; Xu, J.; Zhang, Q.; Chen, F.; Zhang, J.; Gui, K.; Xiong, M.; Li, B.; Ruan, Z.; Zhao, M. OGR1 mediates the inhibitory effects of acidic environment on proliferation and angiogenesis of endothelial progenitor cells. Cell Biol. Int. 2019, 43, 1307–1316. [Google Scholar] [CrossRef] [PubMed]
  64. Othman, F.; Crooks, C.J.; Card, T.R. The risk of Clostridium difficile infection in patients with pernicious anaemia: A retrospective cohort study using primary care database. United European Gastroenterol. J. 2017, 5, 959–966. [Google Scholar] [CrossRef]
  65. Ouyang, S.; Li, Y.; Wu, X.; Wang, Y.; Liu, F.; Zhang, J.; Qiu, Y.; Zhou, Z.; Wang, Z.; Xia, W.; et al. GPR4 signaling is essential for the promotion of acid-mediated angiogenic capacity of endothelial progenitor cells by activating STAT3/VEGFA pathway in patients with coronary artery disease. Stem Cell Res. Ther. 2021, 12, 149. [Google Scholar] [CrossRef] [PubMed]
  66. Ren, J.; Zhang, Y.; Cai, H.; Ma, H.; Zhao, D.; Zhang, X.; Li, Z.; Wang, S.; Wang, J.; Liu, R.; et al. Human GPR4 and the Notch signaling pathway in endothelial cell tube formation. Mol. Med. Rep. 2016, 14, 1235–1240. [Google Scholar] [CrossRef]
  67. Wyder, L.; Suply, T.; Ricoux, B.; Billy, E.; Schnell, C.; Baumgarten, B.U.; Maira, S.M.; Koelbing, C.; Ferretti, M.; Kinzel, B.; et al. Reduced pathological angiogenesis and tumor growth in mice lacking GPR4, a proton sensing receptor. Angiogenesis 2011, 14, 533–544. [Google Scholar] [CrossRef]
  68. Xue, C.; Shao, S.; Yan, Y.; Yang, S.; Bai, S.; Wu, Y.; Zhang, J.; Liu, R.; Ma, H.; Chai, L.; et al. Association between G-protein coupled receptor 4 expression and microvessel density, clinicopathological characteristics and survival in hepatocellular carcinoma. Oncol. Lett. 2020, 19, 2609–2620. [Google Scholar] [CrossRef]
  69. Wenzel, J.; Hansen, C.E.; Bettoni, C.; Vogt, M.A.; Lembrich, B.; Natsagdorj, R.; Huber, G.; Brands, J.; Schmidt, K.; Assmann, J.C.; et al. Impaired endothelium-mediated cerebrovascular reactivity promotes anxiety and respiration disorders in mice. Proc. Natl. Acad. Sci. USA 2020, 117, 1753–1761. [Google Scholar] [CrossRef]
  70. Dong, B.; Zhang, X.; Fan, Y.; Cao, S.; Zhang, X. Acidosis promotes cell apoptosis through the G protein-coupled receptor 4/CCAAT/enhancer-binding protein homologous protein pathway. Oncol. Lett. 2018, 16, 6735–6741. [Google Scholar] [CrossRef] [PubMed]
  71. Dong, L.; Krewson, E.A.; Yang, L.V. Acidosis Activates Endoplasmic Reticulum Stress Pathways through GPR4 in Human Vascular Endothelial Cells. Int. J. Mol. Sci. 2017, 18, 278. [Google Scholar] [CrossRef] [PubMed]
  72. Chen, A.; Dong, L.; Leffler, N.R.; Asch, A.S.; Witte, O.N.; Yang, L.V. Activation of GPR4 by acidosis increases endothelial cell adhesion through the cAMP/Epac pathway. PLoS ONE 2011, 6, e27586. [Google Scholar] [CrossRef]
  73. Weng, Z.; Fluckiger, A.C.; Nisitani, S.; Wahl, M.I.; Le, L.Q.; Hunter, C.A.; Fernal, A.A.; Le Beau, M.M.; Witte, O.N. A DNA damage and stress inducible G protein-coupled receptor blocks cells in G2/M. Proc. Natl. Acad. Sci. USA 1998, 95, 12334–12339. [Google Scholar] [CrossRef] [PubMed]
  74. Wu, M.; Yang, J.P.; Xu, X.Y.; Guo, Z.M.; Bu, B.Y. [Expressions of G2A and OGR1 in peripheral blood cells of patients with hypoxia induced pulmonary hypertension]. Zhongguo Ying Yong Sheng Li Xue Za Zhi 2017, 33, 85–88. [Google Scholar] [CrossRef]
  75. Hattori, T.; Obinata, H.; Ogawa, A.; Kishi, M.; Tatei, K.; Ishikawa, O.; Izumi, T. G2A plays proinflammatory roles in human keratinocytes under oxidative stress as a receptor for 9-hydroxyoctadecadienoic acid. J. Investig. Dermatol. 2008, 128, 1123–1133. [Google Scholar] [CrossRef]
  76. Kern, K.; Schafer, S.M.G.; Cohnen, J.; Pierre, S.; Osthues, T.; Tarighi, N.; Hohmann, S.; Ferreiros, N.; Brune, B.; Weigert, A.; et al. The G2A Receptor Controls Polarization of Macrophage by Determining Their Localization Within the Inflamed Tissue. Front. Immunol. 2018, 9, 2261. [Google Scholar] [CrossRef]
  77. Justus, C.R.; Dong, L.; Yang, L.V. Acidic tumor microenvironment and pH-sensing G protein-coupled receptors. Front. Physiol. 2013, 4, 354. [Google Scholar] [CrossRef]
  78. Chen, P.; Zuo, H.; Xiong, H.; Kolar, M.J.; Chu, Q.; Saghatelian, A.; Siegwart, D.J.; Wan, Y. Gpr132 sensing of lactate mediates tumor-macrophage interplay to promote breast cancer metastasis. Proc. Natl. Acad. Sci. USA 2017, 114, 580–585. [Google Scholar] [CrossRef]
  79. Parks, B.W.; Lusis, A.J.; Kabarowski, J.H. Loss of the lysophosphatidylcholine effector, G2A, ameliorates aortic atherosclerosis in low-density lipoprotein receptor knockout mice. Arterioscler. Thromb. Vasc. Biol. 2006, 26, 2703–2709. [Google Scholar] [CrossRef]
  80. Johnson, L.E.; Elias, M.S.; Bolick, D.T.; Skaflen, M.D.; Green, R.M.; Hedrick, C.C. The G protein-coupled receptor G2A: Involvement in hepatic lipid metabolism and gallstone formation in mice. Hepatology 2008, 48, 1138–1148. [Google Scholar] [CrossRef] [PubMed]
  81. Ishii, S.; Kihara, Y.; Shimizu, T. Identification of T cell death-associated gene 8 (TDAG8) as a novel acid sensing G-protein-coupled receptor. J. Biol. Chem. 2005, 280, 9083–9087. [Google Scholar] [CrossRef] [PubMed]
  82. Tomura, H.; Mogi, C.; Sato, K.; Okajima, F. Proton-sensing and lysolipid-sensitive G-protein-coupled receptors: A novel type of multi-functional receptors. Cell Signal 2005, 17, 1466–1476. [Google Scholar] [CrossRef]
  83. Kyaw, H.; Zeng, Z.; Su, K.; Fan, P.; Shell, B.K.; Carter, K.C.; Li, Y. Cloning, characterization, and mapping of human homolog of mouse T-cell death-associated gene. DNA Cell Biol. 1998, 17, 493–500. [Google Scholar] [CrossRef]
  84. Wang, J.Q.; Kon, J.; Mogi, C.; Tobo, M.; Damirin, A.; Sato, K.; Komachi, M.; Malchinkhuu, E.; Murata, N.; Kimura, T.; et al. TDAG8 is a proton-sensing and psychosine-sensitive G-protein-coupled receptor. J. Biol. Chem. 2004, 279, 45626–45633. [Google Scholar] [CrossRef] [PubMed]
  85. Nagasaka, A.; Mogi, C.; Ono, H.; Nishi, T.; Horii, Y.; Ohba, Y.; Sato, K.; Nakaya, M.; Okajima, F.; Kurose, H. The proton-sensing G protein-coupled receptor T-cell death-associated gene 8 (TDAG8) shows cardioprotective effects against myocardial infarction. Sci. Rep. 2017, 7, 7812. [Google Scholar] [CrossRef]
  86. Tinken, T.M.; Thijssen, D.H.; Hopkins, N.; Dawson, E.A.; Cable, N.T.; Green, D.J. Shear stress mediates endothelial adaptations to exercise training in humans. Hypertension 2010, 55, 312–318. [Google Scholar] [CrossRef]
  87. Tamargo, I.A.; Baek, K.I.; Kim, Y.; Park, C.; Jo, H. Flow-induced reprogramming of endothelial cells in atherosclerosis. Nat. Rev. Cardiol. 2023, 20, 738–753. [Google Scholar] [CrossRef]
  88. Gallardo-Vara, E.; Ntokou, A.; Dave, J.M.; Jovin, D.G.; Saddouk, F.Z.; Greif, D.M. Vascular pathobiology of pulmonary hypertension. J. Heart Lung Transplant. 2023, 42, 544–552. [Google Scholar] [CrossRef]
  89. Green, D.J.; Smith, K.J. Effects of Exercise on Vascular Function, Structure, and Health in Humans. Cold Spring Harb. Perspect. Med. 2018, 8. [Google Scholar] [CrossRef]
  90. Lou, J.; Wu, J.; Feng, M.; Dang, X.; Wu, G.; Yang, H.; Wang, Y.; Li, J.; Zhao, Y.; Shi, C.; et al. Exercise promotes angiogenesis by enhancing endothelial cell fatty acid utilization via liver-derived extracellular vesicle miR-122-5p. J. Sport. Health Sci. 2022, 11, 495–508. [Google Scholar] [CrossRef] [PubMed]
  91. De Ciuceis, C.; Rizzoni, D.; Palatini, P. Microcirculation and Physical Exercise In Hypertension. Hypertension 2023, 80, 730–739. [Google Scholar] [CrossRef] [PubMed]
  92. Santos-Parker, J.R.; LaRocca, T.J.; Seals, D.R. Aerobic exercise and other healthy lifestyle factors that influence vascular aging. Adv. Physiol. Educ. 2014, 38, 296–307. [Google Scholar] [CrossRef]
  93. McDermott, M.M.; Spring, B.; Tian, L.; Treat-Jacobson, D.; Ferrucci, L.; Lloyd-Jones, D.; Zhao, L.; Polonsky, T.; Kibbe, M.R.; Bazzano, L.; et al. Effect of Low-Intensity vs High-Intensity Home-Based Walking Exercise on Walk Distance in Patients With Peripheral Artery Disease: The LITE Randomized Clinical Trial. JAMA 2021, 325, 1266–1276. [Google Scholar] [CrossRef]
  94. Li, Y.; Hanssen, H.; Cordes, M.; Rossmeissl, A.; Endes, S.; Schmidt-Trucksass, A. Aerobic, resistance and combined exercise training on arterial stiffness in normotensive and hypertensive adults: A review. Eur. J. Sport. Sci. 2015, 15, 443–457. [Google Scholar] [CrossRef] [PubMed]
  95. Joyner, M.J. Effect of exercise on arterial compliance. Circulation 2000, 102, 1214–1215. [Google Scholar] [CrossRef]
  96. Okamoto, T.; Min, S.; Sakamaki-Sunaga, M. Arterial compliance and stiffness following low-intensity resistance exercise. Eur. J. Appl. Physiol. 2014, 114, 235–241. [Google Scholar] [CrossRef]
  97. Wang, H.; Zhang, T.; Zhu, W.; Wu, H.; Yan, S. Acute effects of continuous and interval low-intensity exercise on arterial stiffness in healthy young men. Eur. J. Appl. Physiol. 2014, 114, 1385–1392. [Google Scholar] [CrossRef]
  98. Siasos, G.; Athanasiou, D.; Terzis, G.; Stasinaki, A.; Oikonomou, E.; Tsitkanou, S.; Kolokytha, T.; Spengos, K.; Papavassiliou, A.G.; Tousoulis, D. Acute effects of different types of aerobic exercise on endothelial function and arterial stiffness. Eur. J. Prev. Cardiol. 2016, 23, 1565–1572. [Google Scholar] [CrossRef]
  99. Kresnajati, S.; Lin, Y.Y.; Mundel, T.; Bernard, J.R.; Lin, H.F.; Liao, Y.H. Changes in Arterial Stiffness in Response to Various Types of Exercise Modalities: A Narrative Review on Physiological and Endothelial Senescence Perspectives. Cells 2022, 11, 3544. [Google Scholar] [CrossRef]
  100. Kingsley, J.D.; Mayo, X.; Tai, Y.L.; Fennell, C. Arterial Stiffness and Autonomic Modulation After Free-Weight Resistance Exercises in Resistance Trained Individuals. J. Strength. Cond. Res. 2016, 30, 3373–3380. [Google Scholar] [CrossRef] [PubMed]
  101. Lane, A.D.; Yan, H.; Ranadive, S.M.; Kappus, R.M.; Sun, P.; Cook, M.D.; Harvey, I.; Woods, J.; Wilund, K.; Fernhall, B. Sex differences in ventricular-vascular coupling following endurance training. Eur. J. Appl. Physiol. 2014, 114, 2597–2606. [Google Scholar] [CrossRef] [PubMed]
  102. Park, H.Y.; Jung, W.S.; Kim, S.W.; Lim, K. Effects of Interval Training Under Hypoxia on the Autonomic Nervous System and Arterial and Hemorheological Function in Healthy Women. Int. J. Womens Health 2022, 14, 79–90. [Google Scholar] [CrossRef]
  103. Hong, J.; Park, E.; Lee, J.; Lee, Y.; Rooney, B.V.; Park, Y. Exercise training mitigates ER stress and UCP2 deficiency-associated coronary vascular dysfunction in atherosclerosis. Sci. Rep. 2021, 11, 15449. [Google Scholar] [CrossRef]
  104. Zaman, S.; Raj, I.S.; Yang, A.W.H.; Lindner, R.; Denham, J. Exercise training reduces arterial stiffness in women with high blood pressure: A systematic review and meta-analysis. J. Hypertens. 2024, 42, 197–204. [Google Scholar] [CrossRef]
  105. Tomoto, T.; Liu, J.; Tseng, B.Y.; Pasha, E.P.; Cardim, D.; Tarumi, T.; Hynan, L.S.; Munro Cullum, C.; Zhang, R. One-Year Aerobic Exercise Reduced Carotid Arterial Stiffness and Increased Cerebral Blood Flow in Amnestic Mild Cognitive Impairment. J. Alzheimers Dis. 2021, 80, 841–853. [Google Scholar] [CrossRef] [PubMed]
  106. Weiner, D.E.; Liu, C.K.; Miao, S.; Fielding, R.; Katzel, L.I.; Giffuni, J.; Well, A.; Seliger, S.L. Effect of Long-term Exercise Training on Physical Performance and Cardiorespiratory Function in Adults With CKD: A Randomized Controlled Trial. Am. J. Kidney Dis. 2023, 81, 59–66. [Google Scholar] [CrossRef]
  107. DeVallance, E.; Fournier, S.; Lemaster, K.; Moore, C.; Asano, S.; Bonner, D.; Donley, D.; Olfert, I.M.; Chantler, P.D. The effects of resistance exercise training on arterial stiffness in metabolic syndrome. Eur. J. Appl. Physiol. 2016, 116, 899–910. [Google Scholar] [CrossRef]
  108. Kondo, A.; Yamamoto, S.; Nakaki, R.; Shimamura, T.; Hamakubo, T.; Sakai, J.; Kodama, T.; Yoshida, T.; Aburatani, H.; Osawa, T. Extracellular Acidic pH Activates the Sterol Regulatory Element-Binding Protein 2 to Promote Tumor Progression. Cell Rep. 2017, 18, 2228–2242. [Google Scholar] [CrossRef]
  109. Soto, E.; Ortega-Ramirez, A.; Vega, R. Protons as Messengers of Intercellular Communication in the Nervous System. Front. Cell Neurosci. 2018, 12, 342. [Google Scholar] [CrossRef]
  110. Wasserman, K.; Cox, T.A.; Sietsema, K.E. Ventilatory regulation of arterial H(+) (pH) during exercise. Respir. Physiol. Neurobiol. 2014, 190, 142–148. [Google Scholar] [CrossRef] [PubMed]
  111. Kato, T.; Tsukanaka, A.; Harada, T.; Kosaka, M.; Matsui, N. Effect of hypercapnia on changes in blood pH, plasma lactate and ammonia due to exercise. Eur. J. Appl. Physiol. 2005, 95, 400–408. [Google Scholar] [CrossRef]
  112. Helmlinger, G.; Sckell, A.; Dellian, M.; Forbes, N.S.; Jain, R.K. Acid production in glycolysis-impaired tumors provides new insights into tumor metabolism. Clin. Cancer Res. 2002, 8, 1284–1291. [Google Scholar]
  113. Sylow, L.; Kleinert, M.; Richter, E.A.; Jensen, T.E. Exercise-stimulated glucose uptake—Regulation and implications for glycaemic control. Nat. Rev. Endocrinol. 2017, 13, 133–148. [Google Scholar] [CrossRef] [PubMed]
  114. Kampouras, A.; Hatziagorou, E.; Avramidou, V.; Georgopoulou, V.; Kirvassilis, F.; Hebestreit, H.; Tsanakas, J. Ventilation efficiency to exercise in patients with cystic fibrosis. Pediatr. Pulmonol. 2019, 54, 1584–1590. [Google Scholar] [CrossRef]
  115. Agostoni, P.; Emdin, M.; De Martino, F.; Apostolo, A.; Mase, M.; Contini, M.; Carriere, C.; Vignati, C.; Sinagra, G. Roles of periodic breathing and isocapnic buffering period during exercise in heart failure. Eur. J. Prev. Cardiol. 2020, 27, 19–26. [Google Scholar] [CrossRef]
  116. Lindinger, M.I.; Waller, A.P. Physicochemical Analysis of Mixed Venous and Arterial Blood Acid-Base State in Horses at Core Temperature during and after Moderate-Intensity Exercise. Animals 2022, 12, 1875. [Google Scholar] [CrossRef]
  117. Rossiter, H.B. Exercise: Kinetic Considerations for Gas Exchange. Compr. Physiol. 2011, 1, 203–244. [Google Scholar] [CrossRef]
  118. Nielsen, M.S.; Weber, R.E. Antagonistic interaction between oxygenation-linked lactate and CO2 binding to human hemoglobin. Comp. Biochem. Physiol. A Mol. Integr. Physiol. 2007, 146, 429–434. [Google Scholar] [CrossRef]
  119. Boning, D.; Klarholz, C.; Himmelsbach, B.; Hutler, M.; Maassen, N. Extracellular bicarbonate and non-bicarbonate buffering against lactic acid during and after exercise. Eur. J. Appl. Physiol. 2007, 100, 457–467. [Google Scholar] [CrossRef] [PubMed]
  120. Peronnet, F.; Aguilaniu, B. [Pulmonary and alveolar ventilation, gas exchanges and arterial blood gases during ramp exercise]. Rev. Mal. Respir. 2012, 29, 1017–1034. [Google Scholar] [CrossRef] [PubMed]
  121. Brooks, G.A.; Osmond, A.D.; Arevalo, J.A.; Curl, C.C.; Duong, J.J.; Horning, M.A.; Moreno Santillan, D.D.; Leija, R.G. Lactate as a major myokine and exerkine. Nat. Rev. Endocrinol. 2022, 18, 712. [Google Scholar] [CrossRef]
  122. Hall, M.M.; Rajasekaran, S.; Thomsen, T.W.; Peterson, A.R. Lactate: Friend or Foe. PM R. 2016, 8, S8–S15. [Google Scholar] [CrossRef]
  123. Gladden, L.B. Lactate metabolism: A new paradigm for the third millennium. J. Physiol. 2004, 558, 5–30. [Google Scholar] [CrossRef]
  124. Robergs, R.A.; Ghiasvand, F.; Parker, D. Biochemistry of exercise-induced metabolic acidosis. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2004, 287, R502–R516. [Google Scholar] [CrossRef] [PubMed]
  125. Luszczyk, M.; Flis, D.J.; Szadejko, I.; Laskowski, R.; Ziolkowski, W. Excess postexercise oxygen consumption and fat oxidation in recreationally trained men following exercise of equal energy expenditure: Comparisons of spinning and constant endurance exercise. J. Sports Med. Phys. Fitness 2018, 58, 1781–1789. [Google Scholar] [CrossRef]
  126. Keytsman, C.; Hansen, D.; Wens, I.; Eijnde, B.O. Exercise-induced lactate responses in Multiple Sclerosis: A retrospective analysis. NeuroRehabilitation 2019, 45, 99–106. [Google Scholar] [CrossRef]
  127. Lee, S.; Hong, G.; Park, W.; Lee, J.; Kim, N.; Park, H.; Park, J. The effect of short-term creatine intake on blood lactic acid and muscle fatigue measured by accelerometer-based tremor response to acute resistance exercise. Phys. Act. Nutr. 2020, 24, 29–36. [Google Scholar] [CrossRef]
  128. Hebisz, P.; Hebisz, R.; Borkowski, J.; Zaton, M. Time of VO(2)max plateau and post-exercise oxygen consumption during incremental exercise testing in young mountain bike and road cyclists. Physiol. Res. 2018, 67, 711–719. [Google Scholar] [CrossRef]
  129. Vargas-Molina, S.; Martin-Rivera, F.; Bonilla, D.A.; Petro, J.L.; Carbone, L.; Romance, R.; deDiego, M.; Schoenfeld, B.J.; Benitez-Porres, J. Comparison of blood lactate and perceived exertion responses in two matched time-under-tension protocols. PLoS ONE 2020, 15, e0227640. [Google Scholar] [CrossRef] [PubMed]
  130. Jacob, N.; So, I.; Sharma, B.; Marzolini, S.; Tartaglia, M.C.; Oh, P.; Green, R. Effects of High-Intensity Interval Training Protocols on Blood Lactate Levels and Cognition in Healthy Adults: Systematic Review and Meta-Regression. Sports Med. 2023, 53, 977–991. [Google Scholar] [CrossRef] [PubMed]
  131. Lancha Junior, A.H.; Painelli Vde, S.; Saunders, B.; Artioli, G.G. Nutritional Strategies to Modulate Intracellular and Extracellular Buffering Capacity During High-Intensity Exercise. Sports Med. 2015, 45 (Suppl. 1), S71–S81. [Google Scholar] [CrossRef] [PubMed]
  132. Lindinger, M.I.; Heigenhauser, G.J. Effects of gas exchange on acid-base balance. Compr. Physiol. 2012, 2, 2203–2254. [Google Scholar] [CrossRef]
  133. Korzeniewski, B. Contribution of proton leak to oxygen consumption in skeletal muscle during intense exercise is very low despite large contribution at rest. PLoS ONE 2017, 12, e0185991. [Google Scholar] [CrossRef]
  134. Gough, L.A.; Rimmer, S.; Sparks, S.A.; McNaughton, L.R.; Higgins, M.F. Post-exercise Supplementation of Sodium Bicarbonate Improves Acid Base Balance Recovery and Subsequent High-Intensity Boxing Specific Performance. Front. Nutr. 2019, 6, 155. [Google Scholar] [CrossRef]
  135. Fitts, R.H. Cellular mechanisms of muscle fatigue. Physiol. Rev. 1994, 74, 49–94. [Google Scholar] [CrossRef]
  136. Miller, R.G.; Boska, M.D.; Moussavi, R.S.; Carson, P.J.; Weiner, M.W. 31P nuclear magnetic resonance studies of high energy phosphates and pH in human muscle fatigue. Comparison of aerobic and anaerobic exercise. J. Clin. Investig. 1988, 81, 1190–1196. [Google Scholar] [CrossRef]
  137. Huo, R.X.; Li, W.H.; Wu, H.; He, K.X.; Wang, H.; Zhang, S.; Jiang, S.H.; Li, R.K.; Xue, J.L. Transcription factor ONECUT3 regulates HDAC6/HIF-1α activity to promote the Warburg effect and tumor growth in colorectal cancer. Cell Death Dis. 2025, 16, 149. [Google Scholar] [CrossRef]
  138. Semenza, G.L. Hypoxia-inducible factors: Coupling glucose metabolism and redox regulation with induction of the breast cancer stem cell phenotype. EMBO J. 2017, 36, 252–259. [Google Scholar] [CrossRef]
  139. Zhu, Y.F.; Deng, T.J.; Ma, L.; Sun, L.; Hao, Y.C.; Yu, H.X.; Yuan, F.; Tian, Y.M.; Wang, S. Acid-sensing ion channel 1 in nucleus tractus solitarii neurons contributes to the enhanced CO2-stimulated cardiorespiratory effect in spontaneously hypertensive rats. Life Sci. 2024, 351, 122853. [Google Scholar] [CrossRef] [PubMed]
  140. Fu, M.X.; Lv, M.Z.; Guo, J.Y.; Mei, A.H.; Qian, H.; Yang, H.D.; Wu, W.W.; Liu, Z.X.; Zhong, J.X.; Wei, Y.; et al. The clinical significance of T-cell regulation in hypertension treatment. Front. Immunol. 2025, 16, 1550206. [Google Scholar] [CrossRef] [PubMed]
  141. Swietach, P.; Boedtkjer, E.; Pedersen, S.F. How protons pave the way to aggressive cancers. Nat. Rev. Cancer 2023, 23, 825–841. [Google Scholar] [CrossRef]
  142. Wasserman, K.; Beaver, W.L.; Sun, X.G.; Stringer, W.W. Arterial H+ regulation during exercise in humans. Respir. Physiol. Neurobiol. 2011, 178, 191–195. [Google Scholar] [CrossRef]
  143. Sietsema, K.E.; Cooper, D.M.; Perloff, J.K.; Child, J.S.; Rosove, M.H.; Wasserman, K.; Whipp, B.J. Control of ventilation during exercise in patients with central venous-to-systemic arterial shunts. J. Appl. Physiol. 1988, 64, 234–242. [Google Scholar] [CrossRef]
  144. Palmer, B.F.; Clegg, D.J. Respiratory Acidosis and Respiratory Alkalosis: Core Curriculum 2023. Am. J. Kidney Di.s 2023, 82, 347–359. [Google Scholar] [CrossRef]
  145. Flacke, J.P.; Kumar, S.; Kostin, S.; Reusch, H.P.; Ladilov, Y. Acidic preconditioning protects endothelial cells against apoptosis through p38- and Akt-dependent Bcl-xL overexpression. Apoptosis 2009, 14, 90–96. [Google Scholar] [CrossRef] [PubMed]
  146. Dong, L.; Li, Z.; Leffler, N.R.; Asch, A.S.; Chi, J.T.; Yang, L.V. Acidosis activation of the proton-sensing GPR4 receptor stimulates vascular endothelial cell inflammatory responses revealed by transcriptome analysis. PLoS ONE 2013, 8, e61991. [Google Scholar] [CrossRef]
  147. Namkoong, S.; Kim, C.K.; Cho, Y.L.; Kim, J.H.; Lee, H.; Ha, K.S.; Choe, J.; Kim, P.H.; Won, M.H.; Kwon, Y.G.; et al. Forskolin increases angiogenesis through the coordinated cross-talk of PKA-dependent VEGF expression and Epac-mediated PI3K/Akt/eNOS signaling. Cell Signal 2009, 21, 906–915. [Google Scholar] [CrossRef]
  148. Gaynullina, D.K.; Tarasova, O.S.; Shvetsova, A.A.; Borzykh, A.A.; Schubert, R. The Effects of Acidosis on eNOS in the Systemic Vasculature: A Focus on Early Postnatal Ontogenesis. Int. J. Mol. Sci. 2022, 23, 5987. [Google Scholar] [CrossRef]
  149. Foster, S.R.; Hauser, A.S.; Vedel, L.; Strachan, R.T.; Huang, X.P.; Gavin, A.C.; Shah, S.D.; Nayak, A.P.; Haugaard-Kedstrom, L.M.; Penn, R.B.; et al. Discovery of Human Signaling Systems: Pairing Peptides to G Protein-Coupled Receptors. Cell 2019, 179, 895–908 e821. [Google Scholar] [CrossRef] [PubMed]
  150. Maeyashiki, C.; Melhem, H.; Hering, L.; Baebler, K.; Cosin-Roger, J.; Schefer, F.; Weder, B.; Hausmann, M.; Scharl, M.; Rogler, G.; et al. Activation of pH-Sensing Receptor OGR1 (GPR68) Induces ER Stress Via the IRE1alpha/JNK Pathway in an Intestinal Epithelial Cell Model. Sci. Rep. 2020, 10, 1438. [Google Scholar] [CrossRef] [PubMed]
  151. Jing, Z.; Xu, H.; Chen, X.; Zhong, Q.; Huang, J.; Zhang, Y.; Guo, W.; Yang, Z.; Ding, S.; Chen, P.; et al. The Proton-Sensing G-Protein Coupled Receptor GPR4 Promotes Angiogenesis in Head and Neck Cancer. PLoS ONE 2016, 11, e0152789. [Google Scholar] [CrossRef]
  152. Yang, L.V.; Radu, C.G.; Roy, M.; Lee, S.; McLaughlin, J.; Teitell, M.A.; Iruela-Arispe, M.L.; Witte, O.N. Vascular abnormalities in mice deficient for the G protein-coupled receptor GPR4 that functions as a pH sensor. Mol. Cell Biol. 2007, 27, 1334–1347. [Google Scholar] [CrossRef]
  153. Fukuda, H.; Ito, S.; Watari, K.; Mogi, C.; Arisawa, M.; Okajima, F.; Kurose, H.; Shuto, S. Identification of a Potent and Selective GPR4 Antagonist as a Drug Lead for the Treatment of Myocardial Infarction. ACS Med. Chem. Lett. 2016, 7, 493–497. [Google Scholar] [CrossRef]
  154. Velcicky, J.; Miltz, W.; Oberhauser, B.; Orain, D.; Vaupel, A.; Weigand, K.; Dawson King, J.; Littlewood-Evans, A.; Nash, M.; Feifel, R.; et al. Development of Selective, Orally Active GPR4 Antagonists with Modulatory Effects on Nociception, Inflammation, and Angiogenesis. J. Med. Chem. 2017, 60, 3672–3683. [Google Scholar] [CrossRef] [PubMed]
  155. Foster, J.R.; Ueno, S.; Chen, M.X.; Harvey, J.; Dowell, S.J.; Irving, A.J.; Brown, A.J. N-Palmitoylglycine and other N-acylamides activate the lipid receptor G2A/GPR132. Pharmacol. Res. Perspect. 2019, 7, e00542. [Google Scholar] [CrossRef]
  156. Nii, T.; Prabhu, V.V.; Ruvolo, V.; Madhukar, N.; Zhao, R.; Mu, H.; Heese, L.; Nishida, Y.; Kojima, K.; Garnett, M.J.; et al. Imipridone ONC212 activates orphan G protein-coupled receptor GPR132 and integrated stress response in acute myeloid leukemia. Leukemia 2019, 33, 2805–2816. [Google Scholar] [CrossRef]
  157. Murakami, N.; Yokomizo, T.; Okuno, T.; Shimizu, T. G2A is a proton-sensing G-protein-coupled receptor antagonized by lysophosphatidylcholine. J. Biol. Chem. 2004, 279, 42484–42491. [Google Scholar] [CrossRef]
  158. Lahvic, J.L.; Ammerman, M.; Li, P.; Blair, M.C.; Stillman, E.R.; Fast, E.M.; Robertson, A.L.; Christodoulou, C.; Perlin, J.R.; Yang, S.; et al. Specific oxylipins enhance vertebrate hematopoiesis via the receptor GPR132. Proc. Natl. Acad. Sci. USA 2018, 115, 9252–9257. [Google Scholar] [CrossRef]
  159. Yin, H.; Chu, A.; Li, W.; Wang, B.; Shelton, F.; Otero, F.; Nguyen, D.G.; Caldwell, J.S.; Chen, Y.A. Lipid G protein-coupled receptor ligand identification using beta-arrestin PathHunter assay. J. Biol. Chem. 2009, 284, 12328–12338. [Google Scholar] [CrossRef] [PubMed]
  160. Im, D.S.; Heise, C.E.; Nguyen, T.; O’Dowd, B.F.; Lynch, K.R. Identification of a molecular target of psychosine and its role in globoid cell formation. J. Cell Biol. 2001, 153, 429–434. [Google Scholar] [CrossRef] [PubMed]
  161. Mogi, C.; Tobo, M.; Tomura, H.; Murata, N.; He, X.D.; Sato, K.; Kimura, T.; Ishizuka, T.; Sasaki, T.; Sato, T.; et al. Involvement of proton-sensing TDAG8 in extracellular acidification-induced inhibition of proinflammatory cytokine production in peritoneal macrophages. J. Immunol. 2009, 182, 3243–3251. [Google Scholar] [CrossRef] [PubMed]
  162. Huang, X.P.; Karpiak, J.; Kroeze, W.K.; Zhu, H.; Chen, X.; Moy, S.S.; Saddoris, K.A.; Nikolova, V.D.; Farrell, M.S.; Wang, S.; et al. Allosteric ligands for the pharmacologically dark receptors GPR68 and GPR65. Nature 2015, 527, 477–483. [Google Scholar] [CrossRef]
  163. Kleinloog, J.P.D.; Mensink, R.P.; Roodt, J.O.; Thijssen, D.H.J.; Hesselink, M.K.C.; Joris, P.J. Aerobic exercise training improves not only brachial artery flow-mediated vasodilatation but also carotid artery reactivity: A randomized controlled, cross-over trial in older men. Physiol. Rep. 2022, 10, e15395. [Google Scholar] [CrossRef]
  164. Kleiven, O.; Bjorkavoll-Bergseth, M.F.; Omland, T.; Aakre, K.M.; Froysa, V.; Erevik, C.B.; Greve, O.J.; Melberg, T.H.; Auestad, B.; Skadberg, O.; et al. Endurance exercise training volume is not associated with progression of coronary artery calcification. Scand. J. Med. Sci. Sports 2020, 30, 1024–1032. [Google Scholar] [CrossRef]
  165. Alali, M.H.; Lucas, R.A.I.; Junejo, R.T.; Fisher, J.P. Impact of acute dynamic exercise and arterial shear rate modification on radial artery low-flow mediated constriction in young men. Eur. J. Appl. Physiol. 2022, 122, 1885–1895. [Google Scholar] [CrossRef]
  166. Yuan, W.X.; Liu, H.B.; Gao, F.S.; Wang, Y.X.; Qin, K.R. Effects of 8-week swimming training on carotid arterial stiffness and hemodynamics in young overweight adults. Biomed. Eng. Online 2016, 15, 151. [Google Scholar] [CrossRef]
  167. Sakamoto, R.; Katayose, M.; Yamada, Y.; Neki, T.; Kamoda, T.; Tamai, K.; Yamazaki, K.; Iwamoto, E. High-but not moderate-intensity exercise acutely attenuates hypercapnia-induced vasodilation of the internal carotid artery in young men. Eur. J. Appl. Physiol. 2021, 121, 2471–2485. [Google Scholar] [CrossRef]
  168. Badrov, M.B.; Freeman, S.R.; Zokvic, M.A.; Millar, P.J.; McGowan, C.L. Isometric exercise training lowers resting blood pressure and improves local brachial artery flow-mediated dilation equally in men and women. Eur. J. Appl. Physiol. 2016, 116, 1289–1296. [Google Scholar] [CrossRef]
  169. Hasegawa, N.; Fujie, S.; Kurihara, T.; Homma, T.; Sanada, K.; Sato, K.; Hamaoka, T.; Iemitsu, M. Effects of habitual aerobic exercise on the relationship between intramyocellular or extramyocellular lipid content and arterial stiffness. J. Hum. Hypertens. 2016, 30, 606–612. [Google Scholar] [CrossRef] [PubMed]
  170. Robinson, A.T.; Franklin, N.C.; Norkeviciute, E.; Bian, J.T.; Babana, J.C.; Szczurek, M.R.; Phillips, S.A. Improved arterial flow-mediated dilation after exertion involves hydrogen peroxide in overweight and obese adults following aerobic exercise training. J. Hypertens. 2016, 34, 1309–1316. [Google Scholar] [CrossRef]
  171. Landers-Ramos, R.Q.; Corrigan, K.J.; Guth, L.M.; Altom, C.N.; Spangenburg, E.E.; Prior, S.J.; Hagberg, J.M. Short-term exercise training improves flow-mediated dilation and circulating angiogenic cell number in older sedentary adults. Appl. Physiol. Nutr. Metab. 2016, 41, 832–841. [Google Scholar] [CrossRef] [PubMed]
  172. Iwamoto, E.; Bock, J.M.; Casey, D.P. High-Intensity Exercise Enhances Conduit Artery Vascular Function in Older Adults. Med. Sci. Sports Exerc. 2018, 50, 124–130. [Google Scholar] [CrossRef]
  173. Sherman, S.R.; Lefferts, W.K.; Lefferts, E.C.; Grigoriadis, G.; Lima, N.S.; Fernhall, B.; Baynard, T.; Rosenberg, A.J. The effect of aging on carotid artery wall mechanics during maximal resistance exercise. Eur. J. Appl. Physiol. 2022, 122, 2477–2488. [Google Scholar] [CrossRef]
  174. Kingsley, J.D.; Tai, Y.L.; Mayo, X.; Glasgow, A.; Marshall, E. Free-weight resistance exercise on pulse wave reflection and arterial stiffness between sexes in young, resistance-trained adults. Eur. J. Sport. Sci. 2017, 17, 1056–1064. [Google Scholar] [CrossRef]
  175. Choi, Y.; Akazawa, N.; Zempo-Miyaki, A.; Ra, S.G.; Shiraki, H.; Ajisaka, R.; Maeda, S. Acute Effect of High-Intensity Eccentric Exercise on Vascular Endothelial Function in Young Men. J. Strength. Cond. Res. 2016, 30, 2279–2285. [Google Scholar] [CrossRef] [PubMed]
  176. Hasegawa, N.; Fujie, S.; Horii, N.; Miyamoto-Mikami, E.; Tsuji, K.; Uchida, M.; Hamaoka, T.; Tabata, I.; Iemitsu, M. Effects of Different Exercise Modes on Arterial Stiffness and Nitric Oxide Synthesis. Med. Sci. Sports Exerc. 2018, 50, 1177–1185. [Google Scholar] [CrossRef]
  177. Santos, V.; Massuca, L.M.; Angarten, V.; Melo, X.; Pinto, R.; Fernhall, B.; Santa-Clara, H. Arterial Stiffness following Endurance and Resistance Exercise Sessions in Older Patients with Coronary Artery Disease. Int. J. Environ. Res. Public Health 2022, 19. [Google Scholar] [CrossRef]
  178. Campbell, A.K.; Beaumont, A.J.; Hayes, L.; Herbert, P.; Gardner, D.; Ritchie, L.; Sculthorpe, N. Habitual exercise influences carotid artery strain and strain rate, but not cognitive function in healthy middle-aged females. Eur. J. Appl. Physiol. 2023, 123, 1051–1066. [Google Scholar] [CrossRef]
  179. Dias-Santos, E.G.; Farah, B.Q.; Germano-Soares, A.H.; Correia, M.A.; Souza, A.A.; Hora, J.E.J.; Ritti-Dias, R.M.; Andrade-Lima, A. Effects of Exercise Mode on Arterial Stiffness in Symptomatic Peripheral Artery Disease Patients: A Randomized Crossover Clinical Trial. Ann. Vasc. Surg. 2021, 74, 382–388. [Google Scholar] [CrossRef] [PubMed]
  180. Green, D.J.; Marsh, C.E.; Thomas, H.J.; Lester, L.; Scurrah, K.J.; Haynes, A.; Naylor, L.H. Exercise and Artery Function in Twins: Sex Differences in a Cross-Over Trial. Hypertension 2023, 80, 1343–1352. [Google Scholar] [CrossRef] [PubMed]
  181. Soriano-Maldonado, A.; Martinez-Forte, S.; Ferrer-Marquez, M.; Martinez-Rosales, E.; Hernandez-Martinez, A.; Carretero-Ruiz, A.; Villa-Gonzalez, E.; Barranco-Ruiz, Y.; Rodriguez-Perez, M.A.; Torrente-Sanchez, M.J.; et al. Physical Exercise following bariatric surgery in women with Morbid obesity: Study protocol clinical trial (SPIRIT compliant). Medicine 2020, 99, e19427. [Google Scholar] [CrossRef] [PubMed]
  182. Kim, H.K.; Hwang, C.L.; Yoo, J.K.; Hwang, M.H.; Handberg, E.M.; Petersen, J.W.; Nichols, W.W.; Sofianos, S.; Christou, D.D. All-Extremity Exercise Training Improves Arterial Stiffness in Older Adults. Med. Sci. Sports Exerc. 2017, 49, 1404–1411. [Google Scholar] [CrossRef]
  183. Urabe, J.; Ono, K.; Okagawa, J.; Nakayama, Y.; Yamau, R.; Ishikawa, A. Effect of high-intensity interval exercise on renal artery hemodynamics in healthy young adults. J. Sports Med. Phys. Fitness 2023, 63, 129–135. [Google Scholar] [CrossRef]
  184. Ogoh, S.; Washio, T.; Suzuki, K.; Iemitsu, M.; Hashimoto, T.; Iwamoto, E.; Bailey, D.M. Greater increase in internal carotid artery shear rate during aerobic interval compared to continuous exercise in healthy adult men. Physiol. Rep. 2021, 9, e14705. [Google Scholar] [CrossRef]
Figure 1. Proton-sensing GPCR-related signaling pathway. A reduction in extracellular pH enables the activation of GPR68, GPR4, GPR65, and GPR132 receptors located on the cell membrane. The coupling of Gαq proteins can inhibit cell proliferation, migration, and adhesion. The coupling of Gαs proteins has been demonstrated to reduce cell adhesion and inflammation levels. Furthermore, Gαs proteins play a role in inflammatory regulation, with the activation of G12/13 and the inhibition of pro-inflammatory cytokine secretion. This figure was created using BioRender. Abbreviations: PLC: phospholipase C; AC: adenylate cyclase; cAMP: cyclic adenosine monophosphate; Rho: Ras homologous; ROCK: Rho-associated protein kinase; Ras: Rat sarcoma; ERK: extracellular signal-regulated kinase; PKA: protein kinase A; NF-κB: nuclear factor kappa-B.
Figure 1. Proton-sensing GPCR-related signaling pathway. A reduction in extracellular pH enables the activation of GPR68, GPR4, GPR65, and GPR132 receptors located on the cell membrane. The coupling of Gαq proteins can inhibit cell proliferation, migration, and adhesion. The coupling of Gαs proteins has been demonstrated to reduce cell adhesion and inflammation levels. Furthermore, Gαs proteins play a role in inflammatory regulation, with the activation of G12/13 and the inhibition of pro-inflammatory cytokine secretion. This figure was created using BioRender. Abbreviations: PLC: phospholipase C; AC: adenylate cyclase; cAMP: cyclic adenosine monophosphate; Rho: Ras homologous; ROCK: Rho-associated protein kinase; Ras: Rat sarcoma; ERK: extracellular signal-regulated kinase; PKA: protein kinase A; NF-κB: nuclear factor kappa-B.
Biomolecules 15 00813 g001
Figure 2. Proton-sensing GPCRs regulate the mechanism related to arterial function. The diffusion of CO2 from cellular metabolism into the extracellular fluid and bloodstream generates acidic ions, including H+. These ions have the potential to activate proton-sensing GPCRs located on the cell membrane surface. The activation of proton-sensing GPCRs may play a critical role in the regulation of macrophage differentiation, modulation of endothelial dysfunction, neovascularization, and arterial inflammatory responses. This figure was created using BioRender. Abbreviations: NADH: reduced form of nicotinamide-adenine dinucleotide; NAD+: nicotinamide adenine dinucleotide; CO2: carbon dioxide; H+: hydrion; H2O: water molecule; HCO3: bicarbonate radical; NFκB: Nuclear factor-k-gene binding; ATF3: activating transcription factor 3; VCAM-1: vascular cell adhesion molecule-1; ICAM-1: intercellular adhesion molecule-1; TNF-α: tumor necrosis factor-α; HDL: high-density lipoprotein; GTP: guanosine triphosphate; Rho: Ras homologous; GTPase: guanosine triphosphatase; PLC: phospholipase C; PIP2: guanosine triphosphatase; IP3: inositol triphosphate; DAG: diacylglycerol; PKC: protein kinase C; COX: cyclooxygenase; PGI2: prostaglandin-I-2; GRK: G protein-coupled receptor kinase; AC: adenylate cyclase; cAMP: cyclic adenosine monophosphate; PKA: protein kinase A; MAPK: mitogen-activated protein kinase; IL-6: interleukin-6; IL-8: interleukin-8.
Figure 2. Proton-sensing GPCRs regulate the mechanism related to arterial function. The diffusion of CO2 from cellular metabolism into the extracellular fluid and bloodstream generates acidic ions, including H+. These ions have the potential to activate proton-sensing GPCRs located on the cell membrane surface. The activation of proton-sensing GPCRs may play a critical role in the regulation of macrophage differentiation, modulation of endothelial dysfunction, neovascularization, and arterial inflammatory responses. This figure was created using BioRender. Abbreviations: NADH: reduced form of nicotinamide-adenine dinucleotide; NAD+: nicotinamide adenine dinucleotide; CO2: carbon dioxide; H+: hydrion; H2O: water molecule; HCO3: bicarbonate radical; NFκB: Nuclear factor-k-gene binding; ATF3: activating transcription factor 3; VCAM-1: vascular cell adhesion molecule-1; ICAM-1: intercellular adhesion molecule-1; TNF-α: tumor necrosis factor-α; HDL: high-density lipoprotein; GTP: guanosine triphosphate; Rho: Ras homologous; GTPase: guanosine triphosphatase; PLC: phospholipase C; PIP2: guanosine triphosphatase; IP3: inositol triphosphate; DAG: diacylglycerol; PKC: protein kinase C; COX: cyclooxygenase; PGI2: prostaglandin-I-2; GRK: G protein-coupled receptor kinase; AC: adenylate cyclase; cAMP: cyclic adenosine monophosphate; PKA: protein kinase A; MAPK: mitogen-activated protein kinase; IL-6: interleukin-6; IL-8: interleukin-8.
Biomolecules 15 00813 g002
Figure 3. Regulation of acid–base balance during respiration and metabolic stress. During respiration, the lungs eliminate CO2 derived primarily from aerobic metabolism in muscle cells, as well as CO2 generated through the buffering of excess H⁺ by bicarbonate (HCO3⁻) in the blood. Concurrently, erythrocytes facilitate the transport of oxygen to tissue cells. When pulmonary ventilation or metabolic patterns are altered (e.g., during intense exercise or metabolic stress), the production of H⁺ may transiently exceed the capacity of pH buffer systems to neutralize it. This results in the accumulation of H⁺ in the plasma, leading to a decrease in arterial pH (e.g., from 7.4 to 7.2–7.3) and the development of a mild acidotic state. This figure was created using BioRender. Abbreviations: NaHCO3: sodium hydrogen carbonate; HHb: un-ionized hemoglobin; HbO2: oxyhemoglobin; HbNHCOOH: carbamino hemoglobin; HbNH2: aminohemoglobin; KHCO3: potassium bicarbonate; CO2: carbon dioxide; H+: hydrogen ion; H2O: water; HCO3: bicarbonate ion.
Figure 3. Regulation of acid–base balance during respiration and metabolic stress. During respiration, the lungs eliminate CO2 derived primarily from aerobic metabolism in muscle cells, as well as CO2 generated through the buffering of excess H⁺ by bicarbonate (HCO3⁻) in the blood. Concurrently, erythrocytes facilitate the transport of oxygen to tissue cells. When pulmonary ventilation or metabolic patterns are altered (e.g., during intense exercise or metabolic stress), the production of H⁺ may transiently exceed the capacity of pH buffer systems to neutralize it. This results in the accumulation of H⁺ in the plasma, leading to a decrease in arterial pH (e.g., from 7.4 to 7.2–7.3) and the development of a mild acidotic state. This figure was created using BioRender. Abbreviations: NaHCO3: sodium hydrogen carbonate; HHb: un-ionized hemoglobin; HbO2: oxyhemoglobin; HbNHCOOH: carbamino hemoglobin; HbNH2: aminohemoglobin; KHCO3: potassium bicarbonate; CO2: carbon dioxide; H+: hydrogen ion; H2O: water; HCO3: bicarbonate ion.
Biomolecules 15 00813 g003
Figure 4. The effect of exercise on the formation of an acidic microenvironment. The metabolic processes occurring within muscle cells during exercise result in the production of CO2 and lactate. However, these byproducts cannot be eliminated from the body in an expedient manner, which leads to the formation of a weakly acidic microenvironment. This figure was created using BioRender. Abbreviations: PaCO2: carbon dioxide tension.
Figure 4. The effect of exercise on the formation of an acidic microenvironment. The metabolic processes occurring within muscle cells during exercise result in the production of CO2 and lactate. However, these byproducts cannot be eliminated from the body in an expedient manner, which leads to the formation of a weakly acidic microenvironment. This figure was created using BioRender. Abbreviations: PaCO2: carbon dioxide tension.
Biomolecules 15 00813 g004
Figure 5. Possible mechanisms of exercise-mediated modulation of arterial function by the extracellular acidic microenvironment via proton-sensing GPCRs. Modifications in extracellular pH, occurring during and following exercise, have the potential to influence arterial functionality by regulating signal transduction in VECs and VSMCs. These changes are involved in regulating a number of processes, including apoptosis, adhesion, inflammation, arteriolar diastole, and angiogenesis. This figure was created using BioRender. Abbreviations: GTP: guanosine triphosphate; IP3: inositol triphosphate; DAG: diglyceride; sGC: soluble guanylyl cyclase; cGMP: cyclic guanosine monophosphate; MPK-1: mitogen-activated protein kinase phosphatase-1; COX: cyclooxygenase; PGI2: prostaglandin-I-2; Na+/K+-ATPase: sodium-potassium ATPase; PLC: phospholipase C; PIP2: guanosine triphosphatase; IP3: inositol triphosphate; DAG: diglyceride; PDK: phosphoinositide-dependent protein kinase; Calm: calmodulin; PI3K: phosphatidylinositol 3-hydroxy kinase; Akt: protein kinase B; ATP: adenosine triphosphate; AMPK: AMP-activated protein kinase; AC: adenylate cyclase; cAMP: cyclic adenosine monophosphate; PKA: protein kinase A; VCAM-1: vascular cell adhesion molecule-1; ICAM-1: intercellular adhesion molecule-1.
Figure 5. Possible mechanisms of exercise-mediated modulation of arterial function by the extracellular acidic microenvironment via proton-sensing GPCRs. Modifications in extracellular pH, occurring during and following exercise, have the potential to influence arterial functionality by regulating signal transduction in VECs and VSMCs. These changes are involved in regulating a number of processes, including apoptosis, adhesion, inflammation, arteriolar diastole, and angiogenesis. This figure was created using BioRender. Abbreviations: GTP: guanosine triphosphate; IP3: inositol triphosphate; DAG: diglyceride; sGC: soluble guanylyl cyclase; cGMP: cyclic guanosine monophosphate; MPK-1: mitogen-activated protein kinase phosphatase-1; COX: cyclooxygenase; PGI2: prostaglandin-I-2; Na+/K+-ATPase: sodium-potassium ATPase; PLC: phospholipase C; PIP2: guanosine triphosphatase; IP3: inositol triphosphate; DAG: diglyceride; PDK: phosphoinositide-dependent protein kinase; Calm: calmodulin; PI3K: phosphatidylinositol 3-hydroxy kinase; Akt: protein kinase B; ATP: adenosine triphosphate; AMPK: AMP-activated protein kinase; AC: adenylate cyclase; cAMP: cyclic adenosine monophosphate; PKA: protein kinase A; VCAM-1: vascular cell adhesion molecule-1; ICAM-1: intercellular adhesion molecule-1.
Biomolecules 15 00813 g005
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Yu, F.; Jia, D.; Wang, R. Proton-Sensing G Protein-Coupled Receptors and Their Potential Role in Exercise Regulation of Arterial Function. Biomolecules 2025, 15, 813. https://doi.org/10.3390/biom15060813

AMA Style

Yu F, Jia D, Wang R. Proton-Sensing G Protein-Coupled Receptors and Their Potential Role in Exercise Regulation of Arterial Function. Biomolecules. 2025; 15(6):813. https://doi.org/10.3390/biom15060813

Chicago/Turabian Style

Yu, Fengzhi, Dandan Jia, and Ru Wang. 2025. "Proton-Sensing G Protein-Coupled Receptors and Their Potential Role in Exercise Regulation of Arterial Function" Biomolecules 15, no. 6: 813. https://doi.org/10.3390/biom15060813

APA Style

Yu, F., Jia, D., & Wang, R. (2025). Proton-Sensing G Protein-Coupled Receptors and Their Potential Role in Exercise Regulation of Arterial Function. Biomolecules, 15(6), 813. https://doi.org/10.3390/biom15060813

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop