Transient receptor potential channel-dependent myogenic responsiveness in small-sized resistance arteries

Article information

J Exerc Rehabil Vol. 17, No. 1, 4-10, February, 2021
Publication date (electronic) : 2021 February 23
doi : https://doi.org/10.12965/jer.2040836.418
1School of Exercise & Sport Science, College of Natural Sciences, University of Ulsan, Ulsan, Korea
2Department of Physical Education, College of Education, Chung-Ang University, Seoul, Korea
*Corresponding author: Kwang-Seok Hong, https://orcid.org/0000-0003-4691-5807, Department of Physical Education, College of Education, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, Korea, Email: kshong@cau.ac.kr
Received 2020 December 1; Accepted 2020 December 19.

Abstract

It is well documented that the inherent ability of small arteries and arterioles to regulate intraluminal diameter in response to alterations in intravascular pressure determines peripheral vascular resistance and blood flow (termed myogenic response or pressure-induced vasoconstriction/dilation). This autoregulatory property of resistance arteries is primarily originated from mechanosensitive vascular smooth muscle cells (VSMCs). There are diverse biological apparatuses in the plasma membrane of VSMCs that sense mechanical stimuli and generate intracellular signals for the contractility of VSMCs. Although the roles of transient receptor potential (TRP) channels in pressure-induced vasoconstriction are not fully understood to date, TRP channels that are directly activated by mechanical stimuli (e.g., stretch of VSMCs) or indirectly evoked by intracellular molecules (e.g., inositol trisphosphate) provide the major sources of Ca2+ (e.g., Ca2+ influx or release from the sarcoplasmic reticulum) and in turn, evoke vascular reactivity. This review sought to summarize mounting evidence over several decades that the activation of TRP canonical, TRP melastatin, TRP vanilloid, and TRP polycystin channels contributes to myogenic vasoconstriction.

INTRODUCTION

The microcirculation that consists of feed arteries, arterioles, and capillaries contributes to supplying blood to every organ in the body and is involved in the exchange of oxygen, carbon dioxide, nutrients, hormones, and immune cells (Jackson, 2020). Among those vascular beds in the microcirculation, small-sized resistance arteries serve to evoke homeostasis in the cardiovascular system by regulating blood flow and pressure based on their metabolic and physiological demands (Davis and Hill, 1999). The vascular smooth muscle cells (VSMCs) of resistance arteries are empowered with intrinsic properties that maintain blood flow in response to fluctuations in systemic arterial pressure (Hong et al., 2016). Specifically, arterial vasoconstriction and vasodilation occur during increases and decreases in intravascular blood pressure, respectively, which is referred to as the myogenic response or pressure-induced vasoconstriction/dilation (Davis and Hill, 1999). This inherent capacity prevents delicate resistance arteries from exposure to damaging excessive intravascular pressure (Griffin, 2017).

The myogenic reactivity is known to be mechanosensitive-dependent. Mechanical stresses (e.g., stretch of VSMCs oriented circumferentially around resistance artery during elevation in intravascular pressure) sensed by biological machinery in VSMCs stimulate intracellular signaling pathways that lead to pressure-induced vasoconstriction (Hill et al., 2010). Numerous studies over several decades have established biological mechanosensitive apparatus in VSMCs including extracellular matrix, integrins, cytoskeleton, and G protein-coupled receptors (Hill et al., 2016; Hong et al., 2020). Furthermore, mechanical stimuli-mediated activation of ion channels in VSMCs contributes to myogenic vasoconstriction. In particular, transient receptor potential (TRP) ion channels are reported to be directly and indirectly involved in myogenic vasoconstriction as mechanosensors or downstream signal amplifiers (Nemeth et al., 2020; Tykocki et al., 2017). Thus, this study will focus on one of the key determinants, TRP channels, for pressure-induced vasoconstriction in the microcirculation.

TRANSIENT RECEPTOR POTENTIAL CHANNELS

Mounting evidence indicates that TRP channels play a critical role in regulation of membrane potential and Ca2+ signaling of VSMCs and endothelial cells (ECs). TRP channels exist in families typically classified as canonical (TRP canonical, TRPC), TRP vanilloid (TRPV), TRP melastatin (TRPM), TRP ankyrin, mucolipin, and TRP polycystin (TRPP) channels (Montell, 2001). This classification is based on homology of amino acid sequence. Importantly, TRP channels appear to participate in various physiological or pathological situations within the cardiovascular system. For example, TRP channels modulate pacemaker function and contractility of the heart, and their dysfunction causes cardiac hypertrophy, fibrotic disease, and arrhythmias (Earley and Brayden, 2015). In addition, Ca2+-permeable TRP channels in ECs have been implied to be involved in Ca2+ entry-mediated endothelium-derived hyperpolarization and subsequent vasodilation as well as regulation of vascular permeability, angiogenesis, and vascular remodeling (Zhang and Gutterman, 2011). The TRPC1 channel has been identified to show mechanosensitivity in Xenopus oocytes (Maroto et al., 2005). Further, the TRPC3 channel has been demonstrated to participate in membrane depolarization induced by agonist-dependent activation of pyrimidine receptors in pressurized cerebral arteries (Reading et al., 2005). However, subsequent studies have failed to show that TRPC1 or 3 channels are mechanosensitive in VSMCs or that they contribute to myogenic constriction of cerebral arteries (Dietrich et al., 2007; Reading et al., 2005). Therefore, of direct relevance to the current discussion, TRPC6, TRPM4, TRPV4, and TRPP1/2 channels have been chosen, selectively, for further discussion in the following section since they have been implicated in the regulation of myogenic responsiveness.

TRANSIENT RECEPTOR POTENTIAL CANONICAL 6

TRPC6 channel is a non-selective cation channel (permeable to Na+, K+, and Ca2+) which is found in both arteriolar and venous myocytes of diverse vascular beds (Inoue et al., 2006). In studies of human embryonic kidney-293 (HEK-293) cells expressing the channel, TRPC6 channel was thought to exhibit inherent mechanosensitivity when the cells were mechanically stretched by stimulation with either hypoosmotic buffer or negative pressure (Spassova et al., 2006). The fact that antisense oligodeoxynucleotides-dependent inhibition of TRPC6 channel markedly attenuates membrane depolarization and myogenic constriction in cerebral arteriolar myocytes and arteries indirectly implied mechanosensitivity of TRPC6 channel (Welsh et al., 2002). However, these data have been debated and the direct activation of TRPC6 channel has been questioned in subsequent studies (Geffeney et al., 2011; Inoue et al., 2009; Mederos y Schnitzler et al., 2008). More recently, while TRPC6 channel is linked to myogenic vasoconstriction, this channel is identified to be primarily expressed in the cytoplasm of dissociated cerebral VSMCs, but not the plasma membrane surface (Nemeth et al., 2020). These findings imply that TRPC6 channel may not be considered a compelling mechanosensor in cerebral VSMCs. In addition, it has been suggested that the TRPC6 channel cooperates with G protein-coupled receptors (GPCR) to regulate contractility of VSMCs. Consistent with this, earlier studies showed that the activation of α1-adrenergic receptors results in increased TRPC6 channel-mediated currents in VSMCs of portal vein, which is markedly inhibited by antisense oligonucleodtides targeting TRPC6 channel (Inoue et al., 2001). Activation of GPCR leads to phospholipase C-mediated production of diacylglycerol (DAG) and inositol 1,4,5-trisphosphate (IP3), which have been demonstrated to independently or synergistically activate TRPC6 channel (Albert and Large, 2003; Helliwell and Large, 1997; Hofmann et al., 1999; Itsuki et al., 2014). As the angiotensin II type 1 receptor (AT1R), a member of the superfamily of GPCR, has been suggested to be a primary mechanosensor (Blodow et al., 2014; Mederos y Schnitzler et al., 2008; Mederos y Schnitzler et al., 2011; Schleifenbaum et al., 2014; Storch et al., 2012), it is hypothesized that the membrane deformation of arteriolar myocytes evoked by increased intraluminal pressure or stretch elicits the AT1R-mediated production of DAG and/or IP3, which in turn results in TRPC6 channel activation (Fig. 1). This is supported by prior studies showing that the interplay of TRPC6 channel with mechanosensitive AT1R activation gives rise to membrane depolarization and subsequently contributes to pressure-induced vasoconstriction (Mederos y Schnitzler et al., 2008). However, myogenic responsiveness of mice mesenteric arteries with a genetic deficiency of TRPC6 channels is almost identical to that of control arteries (Schleifenbaum et al., 2014), suggesting that cooperation between the AT1R and the TRPC6 channel may not be critical for myogenic constriction in some vascular beds or species. Further, the knockout of TRPC6 channel surprisingly causes increased myogenic constriction that is accompanied by overexpression of TRPC3 channel possibly evoked by compensatory mechanisms against the deficiency in TRPC6 channel (Dietrich et al., 2005).

Fig. 1

An interplay between mechanosensitive GPCRs and TRP channels for myogenic vasoconstriction in cerebral arteries. The GPCRs that are activated by the longitudinal stretch of VSMCs during increases in intravascular pressure stimulate their downstream pathways (i.e., the production of DAG and IP3). These second messengers evoke TRPC6 channel-mediated Ca2+ and then pressure-induced vasoconstriction. Furthermore, IP3 generation from mechanoactivation of the GPCRs or Ca2+ influx via TRPC6 channel results in IP3R-induced Ca2+ release, which contributes to TRPM4 channel-mediated depolarization and Ca2+ entry through VOCC. Along with Ca2+ influx via TRPC6 channel, this Ca2+ mobilization is also responsible for myogenic vasoconstriction. GPCRs, G protein-coupled receptors; TRP, transient receptor potential; AT1R, angiotensin II type 1 receptor; DAG, diacylglycerol; IP3, inositol trisphosphate; IP3R, inositol trisphosphate receptor; P2YR, purinergic receptor; PIP2, phosphatidylinositol bisphosphate; PLC, phospholipase C; SR, sarcoplasmic reticulum; TRPC6, transient receptor potential canonical 6; TRPM4, transient receptor potential melastatin 4; VOCC, voltage-operated Ca2+ channel; VSMC, vascular smooth muscle cells.

TRANSIENT RECEPTOR POTENTIAL MELASTATIN 4

Channels formed by TRPM4 protein are selectively permeable to monovalent cations (e.g., Na+, K+), but not to divalent cations (e.g., Ca2+) (Launay et al., 2002; Nilius et al., 2003). In contrast to TRPC6 channel that functions in a Ca2+-independent manner, while being activated by second messengers (i.e., DAG, IP3), the TRPM4 channel is regulated by the intracellular level of Ca2+. Evidence collected from whole-cell patch clamp measurements has shown that high levels of intracellular Ca2+ are essential to activate TRPM4 channel on HEK-293 cells (EC50=15 μM; Nilius et al., 2003) and intact VSMCs (EC50=10 μM; Earley et al., 2007). Interestingly, it has been observed under inside-out patch clamp conditions that the sensitivity of TRPM4 channel to intracellular Ca2+ is profoundly diminished (EC50=370 μM), indicating that intracellular components are required to regulate the Ca2+ sensitivity of TRPM4 channel (Earley, 2013; Nilius et al., 2005). These investigations and their interpretation are supported by previous studies showing that protein kinase C (PKC) activation and point mutation at PKC phosphorylation sites affect the sensitivity of TRPM4 channel (Nilius et al., 2005). As described above, since higher levels of intracellular Ca2+ are necessary, local Ca2+ signaling events such as Ca2+ sparks (a robust focal Ca2+ increase in the range of 1–100 μM) released from the sarcoplasmic reticulum (SR) are required for activation of TRPM4 channel, rather than global changes in cytoplasmic Ca2+ which are typically in the order of 100–500 nM (Rubart et al., 1996; Zhuge et al., 2004). Consistent with this, TRPM4 channel-mediated cation currents are significantly suppressed by inhibitors of SR calcium transport ATPase (Gonzales and Earley, 2012; Gonzales et al., 2010). In addition, the activity of TRPM4 channel is governed by PKC-dependent trafficking of the channels. Interestingly, PKCδ, but not PKCα and β, has been identified to cause translocation of TRPM4 channel toward cell membrane (Crnich et al., 2010; Garcia et al., 2011).

It has been reported that the TRPM4 channel is functionally significant in the regulation of myogenic constriction in cerebral resistance arterioles. Thus, knockdown of TRPM4 channel expression using antisense oligodeoxynucleotides has been demonstrated to inhibit increased intraluminal pressure-mediated membrane depolarization and myogenic constriction of cerebral arteries (Earley et al., 2004). Further, specific pharmacological inhibition of TRMP4 channel with 9-phenanthrol similarly inhibits myogenic responsiveness (Gonzales et al., 2010). As there is no apparent evidence supporting inherent mechanosensitivity of TRPM4 channel, a link between mechanosensitive GPCR and TRPM4 channel has been assumed (Fig. 1). Regarding this, when the purinergic P2Y4 and P2Y6 receptors are mechanically stimulated, their downstream signaling pathways (i.e., IP3, PKC) influence TRPM4 channel activation, membrane depolarization, and pressure-induced vasoconstriction in cerebral resistance arteries (Brayden et al., 2013; Li et al., 2014). TRPM4 has also been demonstrated to be activated by the mechanoactivation of AT1bR (a member of GPCR family) and then induce pressure-induced vasoconstriction (Pires et al., 2017).

TRANSIENT RECEPTOR POTENTIAL VANILLOID 4

TRPV4 channel shows selectivity for Ca2+ influx in both VSMCs and ECs (Earley et al., 2005; Marrelli et al., 2007). Although extracellular Ca2+ influx is a key determinant for vasoconstriction, TRPV4 channel-mediated Ca2+ entry in arteriolar myocytes has been postulated, surprisingly, to induce vasorelaxation (Earley et al., 2005; Earley et al., 2009). A specific activator of TRPV4 channel, endothelium-derived arachidonic acid metabolite 11,12 epoxyeicosatrienoic acid (11,12-EET), results in Ca2+ influx in cerebral arteriolar myocytes that subsequently leads to the generation of Ca2+ sparks through Ca2+-induced Ca2+ release (CICR). The local and robust Ca2+ release activates large conductance Ca2+-activated K+ channels (BKCa) and evokes hyperpolarization-mediated vasodilation (Earley et al., 2005; Earley et al., 2009). In addition, genetic ablation of TRPV4 channel markedly increases angiotensin II-mediated vasoconstriction of pressurized cerebral arteries (Mercado et al., 2014), suggesting that TRPV4 channel-dependent vasodilation functions as a negative feedback mechanism to presumably prevent exaggerated vasoconstriction. PKC-mediated phosphorylation of the TRPV4 channel allows for Ca2+ entry (termed Ca2+ sparklets) which presumably induces a CICR-dependent vasodilation (Mercado et al., 2014; Navedo et al., 2006). Thus, consistent with previous studies, it is likely that inhibition of TRPV4 channel may potentiate mechanosensitive GPCR (e.g., AT1R, P2Y4R, P2Y6R)-dependent myogenic constriction.

TRANSIENT RECEPTOR POTENTIAL POLYCYSTIN 1/2

TRPP channels are considered as non-selective cation channels for Na+ and Ca2+ (Clapham et al., 2005). TRPP channels are comprised of TRPP1 and TRPP2 channels that have been detected by immunohistochemical staining in human cerebral arteriolar myocytes (Griffin et al., 1997; Torres et al., 2001). Alternative approaches including real-time quantitative polymerase chain reaction and Western blotting have also observed TRPP1 and TRPP2 channels in cultured VSMCs isolated from mouse aorta (Beech et al., 2004; Qian et al., 2003) and rat cerebral resistance arteries (Narayanan et al., 2013). Interestingly, it has been suggested that TRPP1 and TRPP2 channels make different contributions to myogenic constriction of mouse mesenteric arteries (Sharif-Naeini et al., 2009). While knockout of TRPP1 channel impairs pressure-induced vasoconstriction, TRPP2 channel-directed siRNA enhances myogenic responsiveness in mouse mesenteric arteries (Sharif-Naeini et al., 2009). However, in rat cerebral arteries showing 4-fold higher expression of TRPP2 channel than TRPP1 channel, knockdown of TRPP2 channel reduces myogenic reactivity of rat cerebral arteries (Narayanan et al., 2013). Further, hypotonic buffer-mediated membrane stretch of rat cerebral arteriolar myocytes permits TRPP2 channel-mediated influx of cation currents. As the effects of knockdown of TRPP2 channel on pressure-induced vasoconstriction are reportedly disparate in mouse mesenteric and rat cerebral arteries (enhanced vs. reduced myogenic reactivity, respectively), it has been thus suggested that the role of TRPP2 channel in myogenic responsiveness appears different according to the type of vasculature bed and/or species.

CONCLUSIONS

A variety of TRP channels expressed in VSMCs of small-sized resistance arteries play a crucial role in regulating membrane potential and Ca2+ dynamics that are required for VSMC contractile activity and myogenic reactivity. As pressure-induced vasoconstriction and vasodilation contributes to appropriately controlling blood flow in response to moment-to-moment changes in intravascular pressure, it is likely that the dysfunction of TRP channels in VSMCs may result in cardiovascular disorders such as hypertension, vasospasm, or ischemic stroke. However, while there is convincing evidence that TRP channels are significant types of machinery in VSMCs for myogenic responsiveness, it is still debatable that TRP channels act as an independent mechanosensor or downstream signal amplifier for pressure-induced vasoconstriction or vasodilation. Thus, further investigation is needed to decipher the unanswered questions.

ACKNOWLEDGMENTS

This research was supported by the Chung-Ang University Research Grants in 2019.

Notes

CONFLICT OF INTEREST

No potential conflict of interest relevant to this article was reported.

References

Albert, A.P., & Large, W.A.Synergism between inositol phosphates and diacylglycerol on native TRPC6-like channels in rabbit portal vein myocytes. J Physiol, (2003). 552, 789–795.
Beech, D.J., Muraki, K., & Flemming, R.Non-selective cationic channels of smooth muscle and the mammalian homologues of Drosophila TRP. J Physiol, (2004). 559, 685–706.
Blodow, S., Schneider, H., Storch, U., Wizemann, R., Forst, A.L., Gudermann, T., & Mederos y Schnitzler, M.Novel role of mechanosensitive AT1B receptors in myogenic vasoconstriction. Pflugers Arch, (2014). 466, 1343–1353.
Brayden, J.E., Li, Y., & Tavares, M.J.Purinergic receptors regulate myogenic tone in cerebral parenchymal arterioles. J Cereb Blood Flow Metab, (2013). 33, 293–299.
Clapham, D.E., Julius, D., Montell, C., & Schultz, G.International Union of Pharmacology. XLIX. Nomenclature and structure-function relationships of transient receptor potential channels. Pharmacol Rev, (2005). 57, 427–450.
Crnich, R., Amberg, G.C., Leo, M.D., Gonzales, A.L., Tamkun, M.M., Jaggar, J.H., & Earley, S.Vasoconstriction resulting from dynamic membrane trafficking of TRPM4 in vascular smooth muscle cells. Am J Physiol Cell Physiol, (2010). 299, C682–694.
Davis, M.J., & Hill, M.A.Signaling mechanisms underlying the vascular myogenic response. Physiol Rev, (1999). 79, 387–423.
Dietrich, A., Kalwa, H., Storch, U., Mederos y Schnitzler, M., Salanova, B., Pinkenburg, O., Dubrovska, G., Essin, K., Gollasch, M., Birnbaumer, L., & Gudermann, T.Pressure-induced and store-operated cation influx in vascular smooth muscle cells is independent of TRPC1. Pflugers Arch, (2007). 455, 465–477.
Dietrich, A., Mederos, Y.S.M., Gollasch, M., Gross, V., Storch, U., Dubrovska, G., Obst, M., Yildirim, E., Salanova, B., Kalwa, H., Essin, K., Pinkenburg, O., Luft, F.C., Gudermann, T., & Birnbaumer, L.Increased vascular smooth muscle contractility in TRPC6−/− mice. Mol Cell Biol, (2005). 25, 6980–6989.
Earley, S.TRPM4 channels in smooth muscle function. Pflugers Arch, (2013). 465, 1223–1231.
Earley, S., & Brayden, J.E.Transient receptor potential channels in the vasculature. Physiol Rev, (2015). 95, 645–690.
Earley, S., Heppner, T.J., Nelson, M.T., & Brayden, J.E.TRPV4 forms a novel Ca2+ signaling complex with ryanodine receptors and BKCa channels. Circ Res, (2005). 97, 1270–1279.
Earley, S., Pauyo, T., Drapp, R., Tavares, M.J., Liedtke, W., & Brayden, J.E.TRPV4-dependent dilation of peripheral resistance arteries influences arterial pressure. Am J Physiol Heart Circ Physiol, (2009). 297, H1096–1102.
Earley, S., Straub, S.V., & Brayden, J.E.Protein kinase C regulates vascular myogenic tone through activation of TRPM4. Am J Physiol Heart Circ Physiol, (2007). 292, H2613–2622.
Earley, S., Waldron, B.J., & Brayden, J.E.Critical role for transient receptor potential channel TRPM4 in myogenic constriction of cerebral arteries. Circ Res, (2004). 95, 922–929.
Garcia, Z.I., Bruhl, A., Gonzales, A.L., & Earley, S.Basal protein kinase Cdelta activity is required for membrane localization and activity of TRPM4 channels in cerebral artery smooth muscle cells. Channels (Austin), (2011). 5, 210–214.
Geffeney, S.L., Cueva, J.G., Glauser, D.A., Doll, J.C., Lee, T.H., Montoya, M., Karania, S., Garakani, A.M., Pruitt, B.L., & Goodman, M.B.DEG/ENaC but not TRP channels are the major mechanoelectrical transduction channels in a C. elegans nociceptor. Neuron, (2011). 71, 845–857.
Gonzales, A.L., & Earley, S.Endogenous cytosolic Ca2+ buffering is necessary for TRPM4 activity in cerebral artery smooth muscle cells. Cell Calcium, (2012). 51, 82–93.
Gonzales, A.L., Garcia, Z.I., Amberg, G.C., & Earley, S.Pharmacological inhibition of TRPM4 hyperpolarizes vascular smooth muscle. Am J Physiol Cell Physiol, (2010). 299, C1195–1202.
Griffin, K.A.Hypertensive kidney injury and the progression of chronic kidney disease. Hypertension, (2017). 70, 687–694.
Griffin, M.D., Torres, V.E., Grande, J.P., & Kumar, R.Vascular expression of polycystin. J Am Soc Nephrol, (1997). 8, 616–626.
Helliwell, R.M., & Large, W.A.Alpha 1-adrenoceptor activation of a non-selective cation current in rabbit portal vein by 1,2-diacyl-sn-glycerol. J Physiol, (1997). 499, (Pt 2)417–428.
Hill, M.A., Nourian, Z., Ho, I.L., Clifford, P.S., Martinez-Lemus, L., & Meininger, G.A.Small artery elastin distribution and architecture-focus on three dimensional organization. Microcirculation, (2016). 23, 614–620.
Hill, M.A., Yang, Y., Ella, S.R., Davis, M.J., & Braun, A.P.Large conductance, Ca2+-activated K+ channels (BKCa) and arteriolar myogenic signaling. FEBS Lett, (2010). 584, 2033–2042.
Hofmann, T., Obukhov, A.G., Schaefer, M., Harteneck, C., Gudermann, T., & Schultz, G.Direct activation of human TRPC6 and TRPC3 channels by diacylglycerol. Nature, (1999). 397, 259–263.
Hong, K., Zhao, G., Hong, Z., Sun, Z., Yang, Y., Clifford, P.S., Davis, M.J., Meininger, G.A., & Hill, M.A.Mechanical activation of angiotensin II type 1 receptors causes actin remodelling and myogenic responsiveness in skeletal muscle arterioles. J Physiol, (2016). 594, 7027–7047.
Hong, K.S., Kim, K., & Hill, M.A.Regulation of blood flow in small arteries: mechanosensory events underlying myogenic vasoconstriction. J Exerc Rehabil, (2020). 16, 207–215.
Inoue, R., Jensen, L.J., Jian, Z., Shi, J., Hai, L., Lurie, A.I., Henriksen, F.H., Salomonsson, M., Morita, H., Kawarabayashi, Y., Mori, M., Mori, Y., & Ito, Y.Synergistic activation of vascular TRPC6 channel by receptor and mechanical stimulation via phospholipase C/diacylglycerol and phospholipase A2/ω-hydroxylase/20-HETE pathways. Circ Res, (2009). 104, 1399–1409.
Inoue, R., Jensen, L.J., Shi, J., Morita, H., Nishida, M., Honda, A., & Ito, Y.Transient receptor potential channels in cardiovascular function and disease. Circ Res, (2006). 99, 119–131.
Inoue, R., Okada, T., Onoue, H., Hara, Y., Shimizu, S., Naitoh, S., Ito, Y., & Mori, Y.The transient receptor potential protein homologue TRP6 is the essential component of vascular α1-adrenoceptor-activated Ca2+-permeable cation channel. Circ Res, (2001). 88, 325–332.
Itsuki, K., Imai, Y., Hase, H., Okamura, Y., Inoue, R., & Mori, M.X.PLC-mediated PI(4,5)P2 hydrolysis regulates activation and inactivation of TRPC6/7 channels. J Gen Physiol, (2014). 143, 183–201.
Jackson, W.F.Ion channels and the regulation of myogenic tone in peripheral arterioles. Curr Top Membr, (2020). 85, 19–58.
Launay, P., Fleig, A., Perraud, A.L., Scharenberg, A.M., Penner, R., & Kinet, J.P.TRPM4 is a Ca2+-activated nonselective cation channel mediating cell membrane depolarization. Cell, (2002). 109, 397–407.
Li, Y., Baylie, R.L., Tavares, M.J., & Brayden, J.E.TRPM4 channels couple purinergic receptor mechanoactivation and myogenic tone development in cerebral parenchymal arterioles. J Cereb Blood Flow Metab, (2014). 34, 1706–1714.
Maroto, R., Raso, A., Wood, T.G., Kurosky, A., Martinac, B., & Hamill, O.P.TRPC1 forms the stretch-activated cation channel in vertebrate cells. Nat Cell Biol, (2005). 7, 179–185.
Marrelli, S.P., O’Neil, R.G., Brown, R.C., & Bryan, R.M.PLA2 and TRPV4 channels regulate endothelial calcium in cerebral arteries. Am J Physiol Heart Circ Physiol, (2007). 292, H1390–1397.
Mederos y Schnitzler, M., Storch, U., & Gudermann, T.AT1 receptors as mechanosensors. Curr Opin Pharmacol, (2011). 11, 112–116.
Mederos y Schnitzler, M., Storch, U., Meibers, S., Nurwakagari, P., Breit, A., Essin, K., Gollasch, M., & Gudermann, T.Gq-coupled receptors as mechanosensors mediating myogenic vasoconstriction. EMBO J, (2008). 27, 3092–3103.
Mercado, J., Baylie, R., Navedo, M.F., Yuan, C., Scott, J.D., Nelson, M.T., Brayden, J.E., & Santana, L.F.Local control of TRPV4 channels by AKAP150-targeted PKC in arterial smooth muscle. J Gen Physiol, (2014). 143, 559–575.
Montell, C.Physiology, phylogeny, and functions of the TRP superfamily of cation channels. Sci STKE, (2001). 2001, re1.
Narayanan, D., Bulley, S., Leo, M.D., Burris, S.K., Gabrick, K.S., Boop, F.A., & Jaggar, J.H.Smooth muscle cell transient receptor potential polycystin-2 (TRPP2) channels contribute to the myogenic response in cerebral arteries. J Physiol, (2013). 591, 5031–5046.
Navedo, M.F., Amberg, G.C., Nieves, M., Molkentin, J.D., & Santana, L.F.Mechanisms underlying heterogeneous Ca2+ sparklet activity in arterial smooth muscle. J Gen Physiol, (2006). 127, 611–622.
Nemeth, Z., Hildebrandt, E., Ryan, M.J., Granger, J.P., & Drummond, H.A.Pressure-induced constriction of the middle cerebral artery is abolished in TrpC6 knockout mice. Am J Physiol Heart Circ Physiol, (2020). 319, H42–H50.
Nilius, B., Prenen, J., Droogmans, G., Voets, T., Vennekens, R., Freichel, M., Wissenbach, U., & Flockerzi, V.Voltage dependence of the Ca2+-activated cation channel TRPM4. J Biol Chem, (2003). 278, 30813–30820.
Nilius, B., Prenen, J., Tang, J., Wang, C., Owsianik, G., Janssens, A., Voets, T., & Zhu, M.X.Regulation of the Ca2+ sensitivity of the nonselective cation channel TRPM4. J Biol Chem, (2005). 280, 6423–6433.
Pires, P.W., Ko, E.A., Pritchard, H.A.T., Rudokas, M., Yamasaki, E., & Earley, S.The angiotensin II receptor type 1b is the primary sensor of intraluminal pressure in cerebral artery smooth muscle cells. J Physiol, (2017). 595, 4735–4753.
Qian, Q., Li, M., Cai, Y., Ward, C.J., Somlo, S., Harris, P.C., & Torres, V.E.Analysis of the polycystins in aortic vascular smooth muscle cells. J Am Soc Nephrol, (2003). 14, 2280–2287.
Reading, S.A., Earley, S., Waldron, B.J., Welsh, D.G., & Brayden, J.E.TRPC3 mediates pyrimidine receptor-induced depolarization of cerebral arteries. Am J Physiol Heart Circ Physiol, (2005). 288, H2055–2061.
Rubart, M., Patlak, J.B., & Nelson, M.T.Ca2+ currents in cerebral artery smooth muscle cells of rat at physiological Ca2+ concentrations. J Gen Physiol, (1996). 107, 459–472.
Schleifenbaum, J., Kassmann, M., Szijarto, I.A., Hercule, H.C., Tano, J.Y., Weinert, S., Heidenreich, M., Pathan, A.R., Anistan, Y.M., Alenina, N., Rusch, N.J., Bader, M., Jentsch, T.J., & Gollasch, M.Stretch-activation of angiotensin II type 1a receptors contributes to the myogenic response of mouse mesenteric and renal arteries. Circ Res, (2014). 115, 263–272.
Sharif-Naeini, R., Folgering, J.H., Bichet, D., Duprat, F., Lauritzen, I., Arhatte, M., Jodar, M., Dedman, A., Chatelain, F.C., Schulte, U., Retailleau, K., Loufrani, L., Patel, A., Sachs, F., Delmas, P., Peters, D.J., & Honore, E.Polycystin-1 and -2 dosage regulates pressure sensing. Cell, (2009). 139, 587–596.
Spassova, M.A., Hewavitharana, T., Xu, W., Soboloff, J., & Gill, D.L.A common mechanism underlies stretch activation and receptor activation of TRPC6 channels. Proc Natl Acad Sci U S A, (2006). 103, 16586–16591.
Storch, U., Mederos y Schnitzler, M., & Gudermann, T.G protein-mediated stretch reception. Am J Physiol Heart Circ Physiol, (2012). 302, H1241–1249.
Torres, V.E., Cai, Y., Chen, X., Wu, G.Q., Geng, L., Cleghorn, K.A., Johnson, C.M., & Somlo, S.Vascular expression of polycystin-2. J Am Soc Nephrol, (2001). 12, 1–9.
Tykocki, N.R., Boerman, E.M., & Jackson, W.F.Smooth muscle ion channels and regulation of vascular tone in resistance arteries and arterioles. Compr Physiol, (2017). 7, 485–581.
Welsh, D.G., Morielli, A.D., Nelson, M.T., & Brayden, J.E.Transient receptor potential channels regulate myogenic tone of resistance arteries. Circ Res, (2002). 90, 248–250.
Zhang, D.X., & Gutterman, D.D.Transient receptor potential channel activation and endothelium-dependent dilation in the systemic circulation. J Cardiovasc Pharmacol, (2011). 57, 133–139.
Zhuge, R., Fogarty, K.E., Baker, S.P., McCarron, J.G., Tuft, R.A., Lifshitz, L.M., & Walsh, J.V.Ca2+ spark sites in smooth muscle cells are numerous and differ in number of ryanodine receptors, large-conductance K+ channels, and coupling ratio between them. Am J Physiol Cell Physiol, (2004). 287, C1577–1588.

Article information Continued

Fig. 1

An interplay between mechanosensitive GPCRs and TRP channels for myogenic vasoconstriction in cerebral arteries. The GPCRs that are activated by the longitudinal stretch of VSMCs during increases in intravascular pressure stimulate their downstream pathways (i.e., the production of DAG and IP3). These second messengers evoke TRPC6 channel-mediated Ca2+ and then pressure-induced vasoconstriction. Furthermore, IP3 generation from mechanoactivation of the GPCRs or Ca2+ influx via TRPC6 channel results in IP3R-induced Ca2+ release, which contributes to TRPM4 channel-mediated depolarization and Ca2+ entry through VOCC. Along with Ca2+ influx via TRPC6 channel, this Ca2+ mobilization is also responsible for myogenic vasoconstriction. GPCRs, G protein-coupled receptors; TRP, transient receptor potential; AT1R, angiotensin II type 1 receptor; DAG, diacylglycerol; IP3, inositol trisphosphate; IP3R, inositol trisphosphate receptor; P2YR, purinergic receptor; PIP2, phosphatidylinositol bisphosphate; PLC, phospholipase C; SR, sarcoplasmic reticulum; TRPC6, transient receptor potential canonical 6; TRPM4, transient receptor potential melastatin 4; VOCC, voltage-operated Ca2+ channel; VSMC, vascular smooth muscle cells.