Open Access

Roles of small GTPases in cardiac hypertrophy (Review)

  • Authors:
    • Xin Wang
    • Xinwen Nie
    • Hao Wang
    • Zhanhong Ren
  • View Affiliations

  • Published online on: September 19, 2024     https://doi.org/10.3892/mmr.2024.13332
  • Article Number: 208
  • Copyright: © Wang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

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


Abstract

Cardiac hypertrophy results from the heart reacting and adapting to various pathological stimuli and its persistent development is a major contributing factor to heart failure. However, the molecular mechanisms of cardiac hypertrophy remain unclear. Small GTPases in the Ras, Rho, Rab, Arf and Ran subfamilies exhibit GTPase activity and play crucial roles in regulating various cellular responses. Previous studies have shown that Ras, Rho and Rab are closely linked to cardiac hypertrophy and that their overexpression can induce cardiac hypertrophy. Here, we review the functions of small GTPases in cardiac hypertrophy and provide additional insights and references for the prevention and treatment of cardiac hypertrophy.

Introduction

Heart failure leads to increased morbidity and mortality and cardiac hypertrophy is an independent risk factor for the development of heart failure (1,2). Cardiac hypertrophy results from long-term pressure overload and is characterized by the enlargement of myocardial cells and enhanced contractility, which enable the heart to maintain normal blood pumping ability (1,3,4). Chronic concentric hypertrophy can lead to changes in the expression of hypertrophy-related genes such as Nppa, Nppb and myosin heavy chain 7, systolic dysfunction and extracellular remodeling (4,5). Reactivation of Nppa and Nppb can promote sodium excretion, lower blood pressure and ameliorate the burden on the heart, resulting in antihypertrophic effects. Long-term cardiac hypertrophy eventually leads to heart failure (2,6). Studies on the molecular mechanisms of cardiac hypertrophy are beneficial for the prevention and treatment of heart failure.

Previous studies have shown that small GTPases, a known type of molecular switch in cells, play a critical role in cardiac hypertrophy (7,8). They have a molecular weight of only 20–30 kDa and GTPase activity (9). These proteins can be divided into five families based on their amino acid sequence: Ras, Rho, Rab, Arf and Ran (9,10). Since the discovery of the Ras protein in the early 1980s, more than 100 types of small GTPases have been identified (10,11). Small GTPases are in GDP-bound (inactive) and GTP-bound (active) states, which links upstream signaling molecules with downstream effectors to mediate cell proliferation, differentiation, transportation and cytoskeleton regulation (12). Ras mainly regulates cell signaling pathways, including the Ras/mitogen-activated protein kinase kinase kinase (MEKK)/c-Jun N-terminal kinase (JNK) and p38 mitogen-activated protein kinase (MAPK) pathways and plays important roles in cell proliferation, differentiation, morphology and apoptosis (1317). Rho is not only involved in regulating actin, cell polarity, cell migration, vesicle transport and cytokinesis, but is also involved in hematopoiesis, especially in the classical and noncanonical Wnt signaling pathways (1821). Rab is involved in vesicle and endocytic membrane trafficking and can regulate plasma membrane delivery, organelle biogenesis and degradation pathways, lysosomes and autophagy (22). In addition to its basic role in membrane transport, Arf also regulates mitosis, plasma membrane signaling, ciliary transport and lipid droplet functions (23). Ran mainly controls the entry and exit of cargoes between the cytoplasm and nucleus via the nuclear pore complex (24).

Since the discovery of the role of Ras in cardiac hypertrophy in 1993, small GTPases, mainly Ras, Rho and Rab, have been confirmed to be involved in cardiac hypertrophy. Ras can mediate cardiac hypertrophy through multiple pathways, including the MAPK and Ca2+/calcineurin/nuclear factor of activated T cells (NFAT) pathways (14). The Rho A/Rho-associated protein kinase (ROCK) pathway is the main pathway involved in Rho A-mediated cardiac hypertrophy (25). Rac1, a member of the Rho family, can cause cardiac hypertrophy by regulating the MAPK signaling pathway and nicotinamide adenine dinucleotide phosphate (NADPH) oxidase activity (2630). By contrast, Cdc42 can antagonize cardiac hypertrophy by activating JNK and inhibiting calcineurin-NFAT activity (31). At present, Rab1a, Rab4 and Rab4a from the Rab family have been found to lead to myocardial hypertrophy (32). However, the relationships between the other two small GTPases (Arf and Ran) and cardiac hypertrophy remain unreported. The present study reviewed recent research progress on the roles of small GTPases in cardiac hypertrophy, which is helpful for the prevention and treatment of cardiac hypertrophy and heart failure.

Small GTPases in cardiac hypertrophy

Ras family

The Ras family contains 36 members that can be classified into six types: Ras, Ral, Rit, Rap, Rheb and Rad (Ras associated with diabetes) (14). Ras is involved in regulating cell division, cell migration, adhesion, differentiation and apoptosis (Table I) (33). Ral can regulate neuronal plasticity, the immune response and glucose and lipid homeostasis (34). Rit is involved in cell transformation, cell survival, neuromorphogenesis and the regulation of dopamine transporters and plays important roles in autism and schizophrenia (35). Rap plays a role in the normal tissue system, cancer, immune system and hematopoietic system (36). Rheb is highly expressed in the brain, can activate mammalian target of rapamycin complex 1 in response to a variety of growth factor stimuli and is associated with cell growth, protein synthesis and regeneration (37,38). Rad acts as a calcium channel regulator in the normal heart by interacting with cardiac L-type calcium (Table I) (39). Ras and Rad play important roles in cardiac hypertrophy (14,40). However, the relationships between the other four members of the Ras subfamily and cardiac hypertrophy are still unknown.

Table I.

Small GTPases.

Table I.

Small GTPases.

First author/s, yearSmall GTPases Downstream effectorsBasic Function(Refs.)
Tomazini and Shifman, 2023Ras familyRas(JNK, P38, calcineurin, NFAT)Cell division, migration, adhesion, differentiation and apoptosis(33)
Li et al, 2020 RadCaMK IICardiomyocyte calcium signaling(39)
Kilian et al, 2021Rho familyRhoAROCKActin dynamics, signal transduction, gene transcription and cell cycle(60)
Higuchi et al, 2003 Rac1(MEK, ERK, JNK, P38, ASK, NADPH oxidase)Actin dynamics, cell morphology, adhesion and migration(28)
Liu et al, 2022 Cdc42JNK, NAFTActin dynamics, filopodia extension, cell polarity and migration(32)
Filipeanu et al, 2008Rab familyRab1a/Endoplasmic reticulum to Golgi apparatus transport(99)
Filipeanuet al, 2006 Rab4/Endosome to plasma membrane transport(89)
Etzion et al, 2010 Rab4a/Translocation of glucose transporter 4 (GLUT4) and endosomal recycling(100)

[i] /, no reports; JNK, c-Jun N-terminal kinase; NFAT, nuclear factor of activated T cells; ROCK, RhoA/Rho-associated protein kinase; MEK, mitogen-activated protein kinase kinase; ERK, extracellular signal-regulated kinase; ASK, apoptosis signal-regulated kinase; GLUT4, glucose transporter 4.

Ras can induce cardiac hypertrophy (41,42). Microinjection of activated Ras into primary neonatal rat ventricular cardiomyocytes (NRCMs) can upregulate the expression of hypertrophy-related genes, including c-Fos and atrial natriuretic factor (ANF), in vitro (41). In addition, dominant negative mutant Ras (DN-Ras) has an antihypertrophic effect in vitro in NRCMs and plays a protective role in the rat heart in vivo by inhibiting nuclear factor of activated T-cells (NFATs) (42).

Ras can induce cardiac hypertrophy mainly through activating the JNK, p38 MAPK and Ca2+/calcineurin/NFAT signaling pathways both in vitro and in vivo (Fig. 1) (15,17,43,44). The activation of the Ras/MEKK/JNK pathway can mediate ANF gene expression, which causes cardiac hypertrophy both in vitro and in vivo (15). MEKK is a serine triphosphate kinase that can be activated by active Ras in vitro and can regulate stress-activated protein kinases and mitogen-activated protein kinases (45,46). The activation of JNK by MEKK can promote c-Jun transcriptional activity and upregulate ANF gene expression in phenylephrine-treated cardiomyocytes in vitro (15). In activated Ras transgenic mice, JNK activity is upregulated in the left ventricle, which can result in increased ventricular ANF and cardiac hypertrophy in vivo (15). Mechanical stress-activated integrins can trigger autophosphorylation of focal adhesion kinase (FAK) at Tyr397, which can promote the interaction between FAK and c-Src in vitro (17). Tyr925 of FAK is then phosphorylated by c-Src (17). It has been confirmed that the phosphorylation of Tyr397 and Tyr925 in FAK is required for p38 MAPK activation (17). In addition, p38 MAPK activation induced by integrins also requires activated Ras (17). Therefore, it has been suggested that mechanical stress can activate p38 MAPK through the integrin/FAK/Src/Ras pathway to induce cardiomyocyte hypertrophy in vitro (17). In addition, the downregulation of Ras can inhibit the calcineurin activity stimulated by phenylephrine and its upregulation can lead to an increase in the activity of the cytosolic transcription factor NFAT in vitro, which indicates that Ras can activate the Ca2+/calcineurin/NFAT signaling pathway in cardiomyocytes (44). Rad was originally identified in the skeletal muscle of patients with type 2 diabetes (47). Rad is significantly downregulated in pressure overload and phenylephrine-induced cardiac hypertrophy and decreasing Rad can lead to significant enhancement of the phosphorylation and activity of CaMKII, which induces cardiac hypertrophy both in vitro and in vivo (40). Therefore, Rad can antagonize cardiac hypertrophy by inhibiting the activity of CaMKII (40).

Statins or farnesyltransferase inhibitors can suppress Ras by intervening with its farnesylation or geranylgeranylation at its carboxy terminus both in vitro and in vivo (4850). Previous studies have shown that simvastatin can inhibit the progression of left ventricular hypertrophy and that the farnesyltransferase inhibitor FTI-276 can ameliorate cardiac remodeling in experimental animal models (4850). However, it has been proven that statins may cause muscle problems, liver dysfunction, renal insufficiency, poor uptake and short persistence in clinical application (51). A study on farnesyltransferase inhibitors is still at the experimental stage and they have not undergone clinical tests (49). At present, there are fewer effective Ras-targeting drugs with few side effects. Therefore, it is worth developing more Ras-targeting drugs to prevent cardiac hypertrophy.

Rho family

The human Rho family consists of 22 genes encoding >20 proteins (52). According to sequence homology, the Rho family includes the Rho, Rac, Cdc42, RhoD, Rnd and RhoH subfamilies (20). Rho is closely linked to the development of cardiovascular disease through promoting endothelial cell barrier dysfunction (53). Rac is involved in various cellular processes, such as cell division and migration (54). Cdc42 has been shown to play an important role in regulating actin dynamics, filopodia extension, cell polarity and migration (55,56). RhoD and Rnd can both mediate actin cytoskeleton dynamics (57,58). RhoH is beneficial for cell survival, migration and invasion (59). It has been reported that the RhoA, Rac1 and Cdc42 proteins of the Rho family play important roles in cardiac hypertrophy (7,60). However, the relationships between other members of the Rho family and cardiac hypertrophy remain to be elucidated.

RhoA can mediate cardiac hypertrophy through the ROCK signaling pathway (Fig. 2) (25,61,62). The inhibition of RhoA can alleviate cardiac hypertrophy both in vitro and in vivo (6365). ROCK is a downstream molecule of RhoA and RhoA can participate in regulating cardiac hypertrophy by regulating the ROCK signaling pathway (25,61,62). ROCK is divided into two types, ROCK1 and ROCK2, which play different roles in maintaining normal cardiac structure and function (66). A lack of ROCK1 can inhibit cardiac fibrosis and cell apoptosis (67). In addition to cardiac fibrosis and apoptosis, ROCK2 depletion can also ameliorate cardiac hypertrophy (68,69). In addition, the activity of ROCK2 in pressure overload-induced hypertrophied hearts is upregulated, which contributes to cardiac hypertrophy (70). Therefore, it is hypothesized that RhoA may be involved in cardiac hypertrophy through regulating ROCK2. However, further studies are needed to reveal whether RhoA regulates cardiac hypertrophy through ROCK2 and other downstream signaling molecules.

Rac1 can induce cardiac hypertrophy mainly through the MAPK pathway, the apoptosis signal-regulated kinase 1 (ASK1)/NF-κB pathway and NADPH oxidase both in vitro and in vivo (2630,71,72). To reveal the role of Rac1 in cardiac hypertrophy, an activating mutant (V12 Rac1) and a negative mutant (N17 Rac1) were used in a previous in vitro study (71). The overexpression of V12 Rac1 can cause cardiac hypertrophy, while that of N17 Rac1 can inhibit the hypertrophic responses induced by phenylephrine stimulation (71). Previous studies have shown that Rac1 can mediate cardiac hypertrophy by phosphorylating mitogen-activated protein kinase kinase 1/2 (MEK1/2) and extracellular signal-regulated kinase (ERK)1/2 (Fig. 3) both in vitro and in vivo (26,73). The inhibition of the Rac1/JNK pathway can reduce the reactivation of ANF and ameliorate cardiomyocyte hypertrophy in vitro (27,74). Mechanical stress can induce cardiac hypertrophy through the Rac1/reactive oxygen species (ROS)/p38 MAPK pathway in vitro (30). In addition, the overexpression of Rac1 can induce the activation of ASK1 and NF-κB, which leads to cardiomyocyte hypertrophy in vitro (28). ASK1 and NF-κB play important roles in mediating the hypertrophic response of cardiomyocytes (28). NADPH oxidase activity is also closely linked to cardiac hypertrophy induced by Rac1 both in vitro and in vivo (29,75). Rac1 can regulate NADPH oxidase activity through interaction with its components, including gp91phox and p67phox and the upregulation of NADPH oxidase activity can lead to the development of cardiac hypertrophy (29).

In contrast to other small GTPases, Cdc42 can antagonize cardiac hypertrophy in vivo (31). Cdc42 can reduce calcineurin/NFAT activity by stimulating JNK, which leads to cardiac hypertrophy and prevents the development of heart failure (31). Whether there are other signaling pathways through which Cdc2 antagonizes cardiac hypertrophy remains unclear.

The use of Rho-targeting medicines for treating cardiac hypertrophy has been reported. Statins are powerful cardiac protectants that have been found to inhibit cardiac hypertrophy by blocking Rho isopentenylation as inhibitors of 3-hydroxy-3-methylglutaryl-CoA reductase both in vitro and in vivo (76,77). 1,25-Dihydroxyvitamin D3 (VitD) exerts its anti-cardiac hypertrophy effect by downregulating Rac1 in vivo (78). Alendronate, a farnesyl pyrophosphate synthase inhibitor, can inhibit angiotensin (Ang) II-induced neonatal cardiomyocyte hypertrophy by inactivating RhoA both in vitro and in vivo (79). In addition, the RhoA inhibitor Clostridium botulinum C3 exozyme and the ROCK inhibitor Y-27632 exhibit anti-cardiac hypertrophy effects in vitro (80). Glucagon like peptide 1 may reverse cardiac hypertrophy by inhibiting the RhoA/ROCK2 signaling pathway both in vitro and in vivo (81). The selective ROCK inhibitor fasudil can treat various cardiovascular diseases, such as cerebral ischemia and pulmonary hypertension, but its effect on cardiac hypertrophy remains unknown (82,83). The Rac1 inhibitor NSC23766 can antagonize cardiac hypertrophy induced by active Rac1 both in vitro and in vivo (26). There is no Cdc42-targeting medicine for preventing cardiac hypertrophy. The aforementioned Rho-targeting drugs and inhibitors do not undergo clinical tests for treating cardiac hypertrophy and further studies are needed to reveal their detailed roles in ameliorating cardiac hypertrophy.

Rab family

The Rab gene is widely distributed and encodes at least 60 different members of the Rab family in humans (84,85). The Rab family is mainly divided into 10 subfamilies: Rab1, Rab3, Rab4, Rab5, Rab6, Rab8, Rab11, Rab22, Rab27 and Rab40 (85,86). Rab1 is involved in vesicular trafficking from the endoplasmic reticulum to the Golgi apparatus and its dysregulation is associated with the development of cardiomyopathy (87). Rab3 can promote large dense-core vesicle fusion (88). Rab4 can mediate protein transport from the endosome to the plasma membrane (Table I) (89). Rab5 plays a critical role in the early steps of endocytosis (90). Rab6 can regulate not only anterograde transport pathways from the Golgi apparatus but also retrograde trafficking pathways to the Golgi apparatus (91). Rab8 can mediate anterograde membrane trafficking and plays an important role in cell morphogenesis and cell migration (92). Rab11 can regulate the recycling of endosomal cargo proteins to the plasma membrane and is involved in cell migration (93,94). Rab22 plays a critical role in early endosomal recycling (95). Rab27 can promote exocytosis (96). Rab40 can mediate actin dynamics during cell migration (97). In addition, there are some Rab proteins. The roles of Rab1a, Rab4 and Rab4a in cardiac hypertrophy have been reported (89,98,99). However, the relationship between other Rab proteins and cardiac hypertrophy remains unclear.

In transgenic mice overexpressing Rab1a, the endoplasmic reticulum and Golgi apparatus stack significantly enlarged and secretory cardiac natriuretic peptide granules increased, indicating that overexpression of Rab1a can lead to cardiac hypertrophy both in vitro and in vivo (98). Rab4 participates in the transport of proteins from early endosomes to the plasma membrane and promotes the cycle and activation of β-adrenergic receptor (β-AR) (89,100). Cardiac-specific overexpression of Rab4 can enhance β-AR signaling and lead to cardiac hypertrophy both in vitro and in vivo (89). Rab4a participates in glucose transport and induces β-AR recycling to the plasma membrane (99). Increased myocardial Rab4a expression activates β-adrenergic hypersensitivity and leads to cardiac hypertrophy both in vitro and in vivo (99). Other proteins in the Rab family also play different roles in the heart, but their relationship with cardiac hypertrophy remains to be further studied.

Concluding remarks

Cardiac hypertrophy is an irreversible myocardial injury accompanied by myocardial dysfunction and fibrosis due to long-term pathological overload, eventually leading to heart failure (4). As a molecular switch in cell, small GTPases play an important role in the development of cardiac hypertrophy (7,8). The roles of Ras, Rad, RhoA, Rac1, Cdc42, Rab1a, Rab4 and Rab4a in cardiac hypertrophy have been reported (14,31,40,65,71,89,98,99). The roles of Ras, Rad, RhoA, Rac1 and Cdc42 in cardiac hypertrophy are relatively clear, but further studies are needed (14,25,26,31,40). By comparison, the molecular mechanisms of Rab1a, Rab4 and Rab4a in cardiac hypertrophy remain elusive (89,98,99). However, the associations between other small GTPases, including Arf and Ran and cardiac hypertrophy remain unclear. Further studies are needed.

Previous studies have shown that regulators of G-protein signaling can suppress small GTPases by inactivating the heterotrimeric G-proteins of G protein-coupled receptors (GPCRs), which act as GTPase-activating proteins (101103). In addition, it has been reported that RGS2 and RGS4 can ameliorate cardiac hypertrophy by suppressing hypertrophy-related signaling initiated by GPCRs in response to upstream agonists such as angiotensin II and phenylephrine in myocardial cells (102,104106). However, direct evidence showing that RGS can exert protective effects on cardiac hypertrophy through mediating small GTPases is lacking. This topic is worthy of further study.

Currently, statins are used in clinical treatment as powerful cardioprotectants because they inhibit Ras and RhoA (48). It has been reported that Rac1-targeting VitD, RhoA-targeting alendronate and inhibitors of Ras, RhoA and Rac1 can exhibit antihypertrophic effects in the experimental stage, but these agents have not been tested clinically for treating cardiac hypertrophy (26,49,78,79,83). There are no reports on the use of Rad, Cdc42 and Rab-targeting medicines for treating cardiac hypertrophy. Further studies on the roles of small GTPases in cardiac hypertrophy and the development of small GTPase-targeting medicines for treating cardiac hypertrophy are needed. The present study is beneficial for further studies on the molecular mechanisms of cardiac hypertrophy and provided more references for the development of prospective therapeutic targets for cardiac hypertrophy and heart failure.

Acknowledgements

Not applicable.

Funding

The present study was supported by the Natural Science Foundation of Hubei Province (grant no. 2022CFB843), the Special Project on Diabetes and Angiopathy (grant no. 2024TNB04) and Hubei University of Science and Technology School-level Fund (grant no. BK202220).

Availability of data and materials

Not applicable.

Authors' contributions

XW, XN and HW edited the manuscript. ZR conceived, edited and finalized the manuscript. Data authentication is not applicable. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Authors' information

ORCID: Zhanhong Ren, https://orcid.org/0000-0002-3582-008X.

References

1 

Shimizu I and Minamino T: Physiological and pathological cardiac hypertrophy. J Mol Cell Cardiol. 97:245–262. 2016. View Article : Google Scholar : PubMed/NCBI

2 

Tham YK, Bernardo BC, Ooi JY, Weeks KL and McMullen JR: Pathophysiology of cardiac hypertrophy and heart failure: Signaling pathways and novel therapeutic targets. Arch Toxicol. 89:1401–1438. 2015. View Article : Google Scholar : PubMed/NCBI

3 

Nakamura M and Sadoshima J: Mechanisms of physiological and pathological cardiac hypertrophy. Nat Rev Cardiol. 15:387–407. 2018. View Article : Google Scholar : PubMed/NCBI

4 

Samak M, Fatullayev J, Sabashnikov A, Zeriouh M, Schmack B, Farag M, Popov AF, Dohmen PM, Choi YH, Wahlers T and Weymann A: Cardiac Hypertrophy: An introduction to molecular and cellular basis. Med Sci Monit Basic Res. 22:75–79. 2016. View Article : Google Scholar : PubMed/NCBI

5 

Gallo S, Vitacolonna A, Bonzano A, Comoglio P and Crepaldi T: ERK: A key player in the pathophysiology of cardiac hypertrophy. Int J Mol Sci. 20:21642019. View Article : Google Scholar : PubMed/NCBI

6 

Oka T, Akazawa H, Naito AT and Komuro I: Angiogenesis and cardiac hypertrophy: Maintenance of cardiac function and causative roles in heart failure. Circ Res. 114:565–571. 2014. View Article : Google Scholar : PubMed/NCBI

7 

Lezoualc'h F, Métrich M, Hmitou I, Duquesnes N and Morel E: Small GTP-binding proteins and their regulators in cardiac hypertrophy. J Mol Cell Cardiol. 44:623–632. 2008. View Article : Google Scholar : PubMed/NCBI

8 

Clerk A and Sugden PH: Small guanine nucleotide-binding proteins and myocardial hypertrophy. Circ Res. 86:1019–1023. 2000. View Article : Google Scholar : PubMed/NCBI

9 

Matozaki T, Nakanishi H and Takai Y: Small G-protein networks: Their crosstalk and signal cascades. Cell Signal. 12:515–524. 2000. View Article : Google Scholar : PubMed/NCBI

10 

Lundquist EA: Small GTPases. Greenwald I: WormBook; pp. 1–18. 2006, PubMed/NCBI

11 

Wennerberg K, Rossman KL and Der CJ: The Ras superfamily at a glance. J Cell Sci. 118:843–846. 2005. View Article : Google Scholar : PubMed/NCBI

12 

Reiner DJ and Lundquist EA: Small GTPases. WormBook. 2018:1–65. 2018. View Article : Google Scholar : PubMed/NCBI

13 

Karnoub AE and Weinberg RA: Ras oncogenes: Split personalities. Nat Rev Mol Cell Biol. 9:517–531. 2008. View Article : Google Scholar : PubMed/NCBI

14 

Ramos-Kuri M, Meka SH, Salamanca-Buentello F, Hajjar RJ, Lipskaia L and Chemaly ER: Molecules linked to Ras signaling as therapeutic targets in cardiac pathologies. Biol Res. 54:232021. View Article : Google Scholar : PubMed/NCBI

15 

Ramirez MT, Sah VP, Zhao XL, Hunter JJ, Chien KR and Brown JH: The MEKK-JNK pathway is stimulated by alpha1-adrenergic receptor and ras activation and is associated with in vitro and in vivo cardiac hypertrophy. J Biol Chem. 272:14057–14061. 1997. View Article : Google Scholar : PubMed/NCBI

16 

Matsuda T, Jeong JI, Ikeda S, Yamamoto T, Gao S, Babu GJ, Zhai P and Del Re DP: H-Ras isoform mediates protection against pressure overload-induced cardiac dysfunction in part through activation of AKT. Circ Heart Fail. 10:e0036582017. View Article : Google Scholar : PubMed/NCBI

17 

Aikawa R, Nagai T, Kudoh S, Zou Y, Tanaka M, Tamura M, Akazawa H, Takano H, Nagai R and Komuro I: Integrins play a critical role in mechanical stress-induced p38 MAPK activation. Hypertension. 39:233–238. 2022. View Article : Google Scholar

18 

Heasman SJ and Ridley AJ: Mammalian Rho GTPases: New insights into their functions from in vivo studies. Nat Rev Mol Cell Biol. 9:690–701. 2008. View Article : Google Scholar : PubMed/NCBI

19 

Schlessinger K, Hall A and Tolwinski N: Wnt signaling pathways meet Rho GTPases. Genes Dev. 23:265–277. 2009. View Article : Google Scholar : PubMed/NCBI

20 

Mosaddeghzadeh N and Ahmadian MR: The RHO Family GTPases: Mechanisms of regulation and signaling. Cells. 10:18312021. View Article : Google Scholar : PubMed/NCBI

21 

Mackay DJ and Hall A: Rho GTPases. J Biol Chem. 273:20685–20688. 1998. View Article : Google Scholar : PubMed/NCBI

22 

Schwartz SL, Cao C, Pylypenko O, Rak A and Wandinger-Ness A: Rab GTPases at a glance. J Cell Sci. 120:3905–3910. 2007. View Article : Google Scholar : PubMed/NCBI

23 

Jackson CL and Bouvet S: Arfs at a glance. J Cell Sci. 127:4103–4109. 2014.PubMed/NCBI

24 

Yoneda Y, Hieda M, Nagoshi E and Miyamoto Y: Nucleocytoplasmic protein transport and recycling of Ran. Cell Struct Funct. 24:425–433. 1999. View Article : Google Scholar : PubMed/NCBI

25 

Na W, Peng G, Jianping Z, Yanzhong C, Shengjiang G and Li C: RhoA/ROCK may involve in cardiac hypertrophy induced by experimental hyperthyroidism. Toxicol Ind Health. 28:831–839. 2012. View Article : Google Scholar : PubMed/NCBI

26 

Sun Y, Xu C, Jiang Z and Jiang X: DEF6(differentially exprehomolog) exacerbates pathological cardiac hypertrophy via RAC1. Cell Death Dis. 14:4832023. View Article : Google Scholar : PubMed/NCBI

27 

Lin KH, Kumar VB, Shanmugam T, Shibu MA, Chen RJ, Kuo CH, Ho TJ, Padma VV, Yeh YL and Huang CY: miR-145-5p targets paxillin to attenuate angiotensin II-induced pathological cardiac hypertrophy via downregulation of Rac 1, pJNK, p-c-Jun, NFATc3, ANP and by Sirt-1 upregulation. Mol Cell Biochem. 476:3253–3260. 2021. View Article : Google Scholar : PubMed/NCBI

28 

Higuchi Y, Otsu K, Nishida K, Hirotani S, Nakayama H, Yamaguchi O, Hikoso S, Kashiwase K, Takeda T, Watanabe T, et al: The small GTP-binding protein Rac1 induces cardiac myocyte hypertrophy through the activation of apoptosis signal-regulating kinase 1 and nuclear factor-kappa B. J Biol Chem. 278:20770–20777. 2003. View Article : Google Scholar : PubMed/NCBI

29 

Satoh M, Ogita H, Takeshita K, Mukai Y, Kwiatkowski DJ and Liao JK: Requirement of Rac1 in the development of cardiac hypertrophy. Proc Natl Acad Sci USA. 103:7432–7437. 2006. View Article : Google Scholar : PubMed/NCBI

30 

Aikawa R, Nagai T, Tanaka M, Zou Y, Ishihara T, Takano H, Hasegawa H, Akazawa H, Mizukami M, Nagai R and Komuro I: Reactive oxygen species in mechanical stress-induced cardiac hypertrophy. Biochem Biophys Res Commun. 289:901–907. 2001. View Article : Google Scholar : PubMed/NCBI

31 

Maillet M, Lynch JM, Sanna B, York AJ, Zheng Y and Molkentin JD: Cdc42 is an antihypertrophic molecular switch in the mouse heart. J Clin Invest. 119:3079–3088. 2009. View Article : Google Scholar : PubMed/NCBI

32 

Liu J, Zheng X and Wu X: The Rab GTPase in the heart: Pivotal roles in development and disease. Life Sci. 306:1208062022. View Article : Google Scholar : PubMed/NCBI

33 

Tomazini A and Shifman JM: Targeting Ras with protein engineering. Oncotarget. 14:672–687. 2023. View Article : Google Scholar : PubMed/NCBI

34 

Apken LH and Oeckinghaus A: The RAL signaling network: Cancer and beyond. Int Rev Cell Mol Biol. 361:21–105. 2021. View Article : Google Scholar : PubMed/NCBI

35 

Shi GX, Cai W and Andres DA: Rit subfamily small GTPases: Regulators in neuronal differentiation and survival. Cell Signal. 25:2060–2068. 2013. View Article : Google Scholar : PubMed/NCBI

36 

Minato N: Rap G protein signal in normal and disordered lymphohematopoiesis. Exp Cell Res. 319:2323–2328. 2013. View Article : Google Scholar : PubMed/NCBI

37 

Zhong Y, Zhou X, Guan KL and Zhang J: Rheb regulates nuclear mTORC1 activity independent of farnesylation. Cell Chem Biol. 29:1037–1045.e4. 2022. View Article : Google Scholar : PubMed/NCBI

38 

Yang W, Pang D, Chen M, Du C, Jia L, Wang L, He Y, Jiang W, Luo L, Yu Z, et al: Rheb mediates neuronal-activity-induced mitochondrial energetics through mTORC1-independent PDH activation. Dev Cell. 56:811–825.e6. 2021. View Article : Google Scholar : PubMed/NCBI

39 

Li Y, Chang Y, Li X, Li X, Gao J, Zhou Y, Wu F, Bai R, Dong T, Ma S, et al: RAD-Deficient human cardiomyocytes develop hypertrophic cardiomyopathy phenotypes due to calcium dysregulation. Front Cell Dev Biol. 8:5858792020. View Article : Google Scholar : PubMed/NCBI

40 

Chang L, Zhang J, Tseng YH, Xie CQ, Ilany J, Brüning JC, Sun Z, Zhu X, Cui T, Youker KA, et al: Rad GTPase deficiency leads to cardiac hypertrophy. Circulation. 116:2976–2983. 2007. View Article : Google Scholar : PubMed/NCBI

41 

Thorburn A, Thorburn J, Chen SY, Powers S, Shubeita HE, Feramisco JR and Chien KR: HRas-dependent pathways can activate morphological and genetic markers of cardiac muscle cell hypertrophy. J Biol Chem. 268:2244–2249. 1993. View Article : Google Scholar : PubMed/NCBI

42 

Ramos-Kuri M, Rapti K, Mehel H, Zhang S, Dhandapany PS, Liang L, García-Carrancá A, Bobe R, Fischmeister R, Adnot S, et al: Dominant negative Ras attenuates pathological ventricular remodeling in pressure overload cardiac hypertrophy. Biochim Biophys Acta. 1853:2870–2884. 2015. View Article : Google Scholar : PubMed/NCBI

43 

Petrich BG and Wang Y: Stress-activated MAP kinases in cardiac remodeling and heart failure; new insights from transgenic studies. Trends Cardiovasc Med. 14:50–55. 2004. View Article : Google Scholar : PubMed/NCBI

44 

Ichida M and Finkel T: Ras regulates NFAT3 activity in cardiac myocytes. J Biol Chem. 276:3524–3530. 2001. View Article : Google Scholar : PubMed/NCBI

45 

Lange-Carter CA and Johnson GL: Ras-dependent growth factor regulation of MEK kinase in PC12 cells. Science. 265:1458–1461. 1994. View Article : Google Scholar : PubMed/NCBI

46 

Russell M, Lange-Carter CA and Johnson GL: Direct interaction between Ras and the kinase domain of mitogen-activated protein kinase kinase kinase (MEKK1). J Biol Chem. 270:11757–11760. 1995. View Article : Google Scholar : PubMed/NCBI

47 

Reynet C and Kahn CR: Rad: A member of the Ras family overexpressed in muscle of type II diabetic humans. Science. 262:1441–1444. 1993. View Article : Google Scholar : PubMed/NCBI

48 

Cho KJ, Hill MM, Chigurupati S, Du G, Parton RG and Hancock JF: Therapeutic levels of the hydroxmethylglutaryl-coenzyme A reductase inhibitor lovastatin activate ras signaling via phospholipase D2. Mol Cell Biol. 31:1110–1120. 2011. View Article : Google Scholar : PubMed/NCBI

49 

Ding J, Chen YX, Chen Y, Mou Y, Sun XT, Dai DP, Zhao CZ, Yang J, Hu SJ and Guo X: Overexpression of FNTB and the activation of Ras induce hypertrophy and promote apoptosis and autophagic cell death in cardiomyocytes. J Cell Mol Med. 24:8998–9011. 2020. View Article : Google Scholar : PubMed/NCBI

50 

Li X, Han J, Li L, Wang KJ and Hu SJ: Effect of farnesyltransferase inhibition on cardiac remodeling in spontaneously hypertensive rats. Int J Cardiol. 168:3340–3347. 2013. View Article : Google Scholar : PubMed/NCBI

51 

Cai T, Abel L, Langford O, Monaghan G, Aronson JK, Stevens RJ, Lay-Flurrie S, Koshiaris C, McManus RJ, Hobbs FDR and Sheppard JP: Associations between statins and adverse events in primary prevention of cardiovascular disease: Systematic review with pairwise, network and dose-response meta-analyses. BMJ. 374:n15372021. View Article : Google Scholar : PubMed/NCBI

52 

Jaiswal M, Dvorsky R and Ahmadian MR: Deciphering the molecular and functional basis of Dbl family proteins: A novel systematic approach toward classification of selective activation of the Rho family proteins. J Biol Chem. 288:4486–4500. 2013. View Article : Google Scholar : PubMed/NCBI

53 

Strassheim D, Gerasimovskaya E, Irwin D, Dempsey EC, Stenmark K and Karoor V: RhoGTPase in vascular disease. Cells. 8:5512019. View Article : Google Scholar : PubMed/NCBI

54 

Lee CF, Carley RE, Butler CA and Morrison AR: Rac GTPase signaling in immune-mediated mechanisms of atherosclerosis. Cells. 10:28082021. View Article : Google Scholar : PubMed/NCBI

55 

Nguyen DT, Gao L, Wong A and Chen CS: Cdc42 regulates branching in angiogenic sprouting in vitro. Microcirculation. 24:10.1111/micc.12372. 2017. View Article : Google Scholar : PubMed/NCBI

56 

Lv J, Zeng J, Guo F, Li Y, Xu M, Cheng Y, Zhang L, Cai S, Chen Y, Zheng Y and Hu G: Endothelial Cdc42 deficiency impairs endothelial regeneration and vascular repair after inflammatory vascular injury. Respir Res. 19:272018. View Article : Google Scholar : PubMed/NCBI

57 

Basbous S, Azzarelli R, Pacary E and Moreau V: Pathophysiological functions of Rnd proteins. Small GTPases. 12:336–357. 2021. View Article : Google Scholar : PubMed/NCBI

58 

Blom M, Reis K and Aspenström P: RhoD localization and function is dependent on its GTP/GDP-bound state and unique N-terminal motif. Eur J Cell Biol. 97:393–401. 2018. View Article : Google Scholar : PubMed/NCBI

59 

Ahmad Mokhtar AM, Hashim IF, Mohd Zaini Makhtar M, Salikin NH and Amin-Nordin S: The Role of RhoH in TCR signalling and its involvement in diseases. Cells. 10:9502021. View Article : Google Scholar : PubMed/NCBI

60 

Kilian LS, Voran J, Frank D and Rangrez AY: RhoA: A dubious molecule in cardiac pathophysiology. J Biomed Sci. 28:332021. View Article : Google Scholar : PubMed/NCBI

61 

Miyamoto S, Del Re DP, Xiang SY, Zhao X, Florholmen G and Brown JH: Revisited and revised: is RhoA always a villain in cardiac pathophysiology? J Cardiovasc Transl Res. 3:330–343. 2010. View Article : Google Scholar : PubMed/NCBI

62 

Zhou Q, Wei SS, Wang H, Wang Q, Li W, Li G, Hou JW, Chen XM, Chen J, Xu WP, et al: Crucial Role of ROCK2-Mediated phosphorylation and upregulation of FHOD3 in the pathogenesis of angiotensin II-induced cardiac hypertrophy. Hypertension. 69:1070–1083. 2017. View Article : Google Scholar : PubMed/NCBI

63 

Sakaguchi T, Takefuji M, Wettschureck N, Hamaguchi T, Amano M, Kato K, Tsuda T, Eguchi S, Ishihama S, Mori Y, et al: Protein Kinase N promotes stress-induced cardiac dysfunction through phosphorylation of myocardin-related transcription factor A and disruption of its interaction with actin. Circulation. 140:1737–1752. 2019. View Article : Google Scholar : PubMed/NCBI

64 

Huang J, Qu Q, Dai Y, Ren D, Qian J and Ge J: Detrimental Role of PDZ-RhoGEF in pathological cardiac hypertrophy. Hypertension. 80:403–415. 2023. View Article : Google Scholar : PubMed/NCBI

65 

Aoki H, Izumo S and Sadoshima J: Angiotensin II activates RhoA in cardiac myocytes: A critical role of RhoA in angiotensin II-induced premyofibril formation. Circ Res. 82:666–676. 1998. View Article : Google Scholar : PubMed/NCBI

66 

Nakagawa O, Fujisawa K, Ishizaki T, Saito Y, Nakao K and Narumiya S: ROCK-I and ROCK-II, two isoforms of Rho-associated coiled-coil forming protein serine/threonine kinase in mice. FEBS Lett. 392:189–193. 1996. View Article : Google Scholar : PubMed/NCBI

67 

Zhang YM, Bo J, Taffet GE, Chang J, Shi J, Reddy AK, Michael LH, Schneider MD, Entman ML, Schwartz RJ and Wei L: Targeted deletion of ROCK1 protects the heart against pressure overload by inhibiting reactive fibrosis. FASEB J. 20:916–925. 2006. View Article : Google Scholar : PubMed/NCBI

68 

Okamoto R, Li Y, Noma K, Hiroi Y, Liu PY, Taniguchi M, Ito M and Liao JK: FHL2 prevents cardiac hypertrophy in mice with cardiac-specific deletion of ROCK2. FASEB J. 27:1439–1449. 2013. View Article : Google Scholar : PubMed/NCBI

69 

Shimizu T, Narang N, Chen P, Yu B, Knapp M, Janardanan J, Blair J and Liao JK: Fibroblast deletion of ROCK2 attenuates cardiac hypertrophy, fibrosis and diastolic dysfunction. JCI Insight. 2:e931872017. View Article : Google Scholar : PubMed/NCBI

70 

Ikeda S, Satoh K, Kikuchi N, Miyata S, Suzuki K, Omura J, Shimizu T, Kobayashi K, Kobayashi K, Fukumoto Y, et al: Crucial role of rho-kinase in pressure overload-induced right ventricular hypertrophy and dysfunction in mice. Arterioscler Thromb Vasc Biol. 34:1260–1271. 2014. View Article : Google Scholar : PubMed/NCBI

71 

Pracyk JB, Tanaka K, Hegland DD, Kim KS, Sethi R, Rovira II, Blazina DR, Lee L, Bruder JT, Kovesdi I, et al: A requirement for the rac1 GTPase in the signal transduction pathway leading to cardiac myocyte hypertrophy. J Clin Invest. 102:929–937. 1998. View Article : Google Scholar : PubMed/NCBI

72 

Elnakish MT, Moldovan L, Khan M, Hassanain HH and Janssen PM: Myocardial Rac1 exhibits partial involvement in thyroxin-induced cardiomyocyte hypertrophy and its inhibition is not sufficient to improve cardiac dysfunction or contractile abnormalities in mouse papillary muscles. J Cardiovasc Pharmacol. 61:536–544. 2013. View Article : Google Scholar : PubMed/NCBI

73 

Li PL, Liu H, Chen GP, Li L, Shi HJ, Nie HY, Liu Z, Hu YF, Yang J, Zhang P, et al: STEAP3 (Six-Transmembrane Epithelial Antigen of Prostate 3) Inhibits Pathological Cardiac Hypertrophy. Hypertension. 76:1219–1230. 2020. View Article : Google Scholar : PubMed/NCBI

74 

Clerk A, Pham FH, Fuller SJ, Sahai E, Aktories K, Marais R, Marshall C and Sugden PH: Regulation of mitogen-activated protein kinases in cardiac myocytes through the small G protein Rac1. Mol Cell Biol. 21:1173–1184. 2001. View Article : Google Scholar : PubMed/NCBI

75 

Sawada N, Li Y and Liao JK: Novel aspects of the roles of Rac1 GTPase in the cardiovascular system. Curr Opin Pharmacol. 10:116–121. 2010. View Article : Google Scholar : PubMed/NCBI

76 

Cacciapuoti F: Molecular mechanisms of left ventricular hypertrophy (LVH) in systemic hypertension (SH)-possible therapeutic perspectives. J Am Soc Hypertens. 5:449–455. 2011. View Article : Google Scholar : PubMed/NCBI

77 

Hauck L, Harms C, Grothe D, An J, Gertz K, Kronenberg G, Dietz R, Endres M and von Harsdorf R: Critical role for FoxO3a-dependent regulation of p21CIP1/WAF1 in response to statin signaling in cardiac myocytes. Circ Res. 100:50–60. 2007. View Article : Google Scholar : PubMed/NCBI

78 

Moradi A, Maroofi A, Hemati M, Hashemzade T, Alborzi N and Safari F: Inhibition of GTPase Rac1 expression by vitamin D mitigates pressure overload-induced cardiac hypertrophy. Int J Cardiol Heart Vasc. 37:1009222021.PubMed/NCBI

79 

Zhang C, Jin DD, Wang XY, Lou L and Yang J: Key enzymes for the mevalonate pathway in the cardiovascular system. J Cardiovasc Pharmacol. 77:142–152. 2021. View Article : Google Scholar : PubMed/NCBI

80 

Zeidan A, Gan XT, Thomas A and Karmazyn M: Prevention of RhoA activation and cofilin-mediated actin polymerization mediates the antihypertrophic effect of adenosine receptor agonists in angiotensin II- and endothelin-1-treated cardiomyocytes. Mol Cell Biochem. 385:239–248. 2014. View Article : Google Scholar : PubMed/NCBI

81 

Fan S, Xiong Q, Zhang X, Zhang L and Shi Y: Glucagon-like peptide 1 reverses myocardial hypertrophy through cAMP/PKA/RhoA/ROCK2 signaling. Acta Biochim Biophys Sin (Shanghai). 52:612–619. 2020. View Article : Google Scholar : PubMed/NCBI

82 

Tawara S and Shimokawa H: Progress of the study of rho-kinase and future perspective of the inhibitor. Yakugaku Zasshi. 127:501–514. 2007. View Article : Google Scholar : PubMed/NCBI

83 

Zhang Y and Wu S: Effects of fasudil on pulmonary hypertension in clinical practice. Pulm Pharmacol Ther. 46:54–63. 2017. View Article : Google Scholar : PubMed/NCBI

84 

Bock JB, Matern HT, Peden AA and Scheller RH: A genomic perspective on membrane compartment organization. Nature. 409:839–841. 2001. View Article : Google Scholar : PubMed/NCBI

85 

Stenmark H and Olkkonen VM: The Rab GTPase family. Genome Biol. 2:REVIEWS30072001. View Article : Google Scholar : PubMed/NCBI

86 

Pereira-Leal JB and Seabra MC: The mammalian Rab family of small GTPases: Definition of family and subfamily sequence motifs suggests a mechanism for functional specificity in the Ras superfamily. J Mol Biol. 301:1077–1087. 2000. View Article : Google Scholar : PubMed/NCBI

87 

Yang XZ, Li XX, Zhang YJ, Rodriguez-Rodriguez L, Xiang MQ, Wang HY and Zheng XF: Rab1 in cell signaling, cancer and other diseases. Oncogene. 35:5699–5704. 2016. View Article : Google Scholar : PubMed/NCBI

88 

Schonn JS, van Weering JR, Mohrmann R, Schlüter OM, Südhof TC, de Wit H, Verhage M and Sørensen JB: Rab3 proteins involved in vesicle biogenesis and priming in embryonic mouse chromaffin cells. Traffic. 11:1415–1428. 2010. View Article : Google Scholar : PubMed/NCBI

89 

Filipeanu CM, Zhou F, Lam ML, Kerut KE, Claycomb WC and Wu G: Enhancement of the recycling and activation of beta-adrenergic receptor by Rab4 GTPase in cardiac myocytes. J Biol Chem. 281:11097–11103. 2006. View Article : Google Scholar : PubMed/NCBI

90 

Xu W, Fang F, Ding J and Wu C: Dysregulation of Rab5-mediated endocytic pathways in Alzheimer's disease. Traffic. 19:253–262. 2018. View Article : Google Scholar : PubMed/NCBI

91 

Dornan LG and Simpson JC: Rab6-mediated retrograde trafficking from the Golgi: The trouble with tubules. Small GTPases. 14:26–44. 2023. View Article : Google Scholar : PubMed/NCBI

92 

Stypulkowski E, Feng Q, Joseph I, Farrell V, Flores J, Yu S, Sakamori R, Sun J, Bandyopadhyay S, Das S, et al: Rab8 attenuates Wnt signaling and is required for mesenchymal differentiation into adipocytes. J Biol Chem. 296:1004882021. View Article : Google Scholar : PubMed/NCBI

93 

Wilson B, Flett C, Gemperle J, Lawless C, Hartshorn M, Hinde E, Harrison T, Chastney M, Taylor S, Allen J, et al: Proximity labelling identifies pro-migratory endocytic recycling cargo and machinery of the Rab4 and Rab11 families. J Cell Sci. 136:jcs2604682023. View Article : Google Scholar : PubMed/NCBI

94 

Banworth MJ and Li G: Consequences of Rab GTPase dysfunction in genetic or acquired human diseases. Small GTPases. 9:158–181. 2018. View Article : Google Scholar : PubMed/NCBI

95 

Banworth MJ, Liang Z and Li G: A novel membrane targeting domain mediates the endosomal or Golgi localization specificity of small GTPases Rab22 and Rab31. J Biol Chem. 298:1022812022. View Article : Google Scholar : PubMed/NCBI

96 

Izumi T: In vivo Roles of Rab27 and its effectors in exocytosis. Cell Struct Funct. 46:79–94. 2021. View Article : Google Scholar : PubMed/NCBI

97 

Neumann AJ and Prekeris R: A Rab-bit hole: Rab40 GTPases as new regulators of the actin cytoskeleton and cell migration. Front Cell Dev Biol. 11:12689222023. View Article : Google Scholar : PubMed/NCBI

98 

Moore I, Schell J and Palme K: Subclass-specific sequence motifs identified in Rab GTPases. Trends Biochem Sci. 20:10–12. 1995. View Article : Google Scholar : PubMed/NCBI

99 

Filipeanu CM, Zhou F and Wu G: Analysis of Rab1 function in cardiomyocyte growth. Methods Enzymol. 438:217–226. 2008. View Article : Google Scholar : PubMed/NCBI

100 

Etzion S, Etzion Y, DeBosch B, Crawford PA and Muslin AJ: Akt2 deficiency promotes cardiac induction of Rab4a and myocardial β-adrenergic hypersensitivity. J Mol Cell Cardiol. 49:931–940. 2010. View Article : Google Scholar : PubMed/NCBI

101 

Seachrist JL and Ferguson SS: Regulation of G protein-coupled receptor endocytosis and trafficking by Rab GTPases. Life Sci. 74:225–235. 2003. View Article : Google Scholar : PubMed/NCBI

102 

Del Calvo G, Baggio Lopez T and Lymperopoulos A: The therapeutic potential of targeting cardiac RGS4. Ther Adv Cardiovasc Dis. 17:175394472311993502023. View Article : Google Scholar : PubMed/NCBI

103 

Lymperopoulos A, Borges JI and Stoicovy RA: RGS proteins and cardiovascular Angiotensin II Signaling: Novel opportunities for therapeutic targeting. Biochem Pharmacol. 218:1159042023. View Article : Google Scholar : PubMed/NCBI

104 

Magalhaes AC, Dunn H and Ferguson SS: Regulation of GPCR activity, trafficking and localization by GPCR-interacting proteins. Br J Pharmacol. 165:1717–1736. 2012. View Article : Google Scholar : PubMed/NCBI

105 

Rogers JH, Tamirisa P, Kovacs A, Weinheimer C, Courtois M, Blumer KJ, Kelly DP and Muslin AJ: RGS4 causes increased mortality and reduced cardiac hypertrophy in response to pressure overload. J Clin Invest. 104:567–576. 1999. View Article : Google Scholar : PubMed/NCBI

106 

Chidiac P, Sobiesiak AJ, Lee KN, Gros R and Nguyen CH: The eIF2B-interacting domain of RGS2 protects against GPCR agonist-induced hypertrophy in neonatal rat cardiomyocytes. Cell Signal. 26:1226–1234. 2014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2024
Volume 30 Issue 5

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wang X, Nie X, Wang H and Ren Z: Roles of small GTPases in cardiac hypertrophy (Review). Mol Med Rep 30: 208, 2024.
APA
Wang, X., Nie, X., Wang, H., & Ren, Z. (2024). Roles of small GTPases in cardiac hypertrophy (Review). Molecular Medicine Reports, 30, 208. https://doi.org/10.3892/mmr.2024.13332
MLA
Wang, X., Nie, X., Wang, H., Ren, Z."Roles of small GTPases in cardiac hypertrophy (Review)". Molecular Medicine Reports 30.5 (2024): 208.
Chicago
Wang, X., Nie, X., Wang, H., Ren, Z."Roles of small GTPases in cardiac hypertrophy (Review)". Molecular Medicine Reports 30, no. 5 (2024): 208. https://doi.org/10.3892/mmr.2024.13332