Role and targeting of the AGC kinase family in pulmonary fibrosis (Review)

  • Authors:
    • Chao Mei
    • Tao Chen
    • Xiangfei Huang
    • Chenlu Xiong
    • Shibiao Chen
    • Yong Li
  • View Affiliations

  • Published online on: March 8, 2024     https://doi.org/10.3892/etm.2024.12478
  • Article Number: 190
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Pulmonary fibrosis (PF) is a progressive and irreversible pulmonary disease with a high mortality rate and limited treatment options. The cAMP‑dependent protein kinase A, cGMP‑dependent protein kinase G and phospholipid‑dependent protein kinase C, collectively known as AGC kinases, are evolutionarily conserved protein kinases that are widely distributed among eukaryotes. AGC kinases serve a crucial role in a variety of cellular functions and pathological processes, including cancer, diabetes, inflammation and viral infections, where they have been implicated the pathogenesis of PF. The present review summarizes the evidence for the involvement of specific AGC kinases in the pathogenesis of PF, and provides a theoretical basis for the development of targeted AGC kinase small molecule inhibitors or targeted drugs, offering more effective treatment options and strategies for patients with PF.

1. Introduction

Pulmonary fibrosis (PF) is a respiratory disease that is characterized by scarring in the lungs and subsequent breathing difficulties (1). There are various types of PF including asbestosis, hypersensitivity pneumonitis and idiopathic pulmonary fibrosis (IPF), with IPF being one of the most common and severe forms (2). IPF is an interstitial lung disease of unknown etiology that is chronic, progressive, and irreversible (3). It is distinguished by epithelial cell activation and injury, fibroblast proliferation and differentiation, extracellular matrix (ECM) deposition, irreversible destruction of the alveolar structure and respiratory insufficiency (Fig. 1) (4). IPF primarily occurs among the middle-aged and elderly populations, where it is limited to the lungs (5,6). The treatment of IPF typically involves a combination of medications, pulmonary rehabilitation and, in some cases, lung transplantation (7). IPF is a chronic and progressive disease, and existing treatments, including antifibrotic medications, aim to slow down the progression rather than cure the condition (7). However, the antifibrotic medications may have side effects, and not all individuals with IPF can tolerate these drugs (8). The causes and pathogenesis of IPF remain unclear and the effects conferred by currently available therapeutic methods are limited (9,10). The survival rate after IPF diagnosis is typically only 2-5 years, where the prognosis of which is even worse compared with that of several types of cancer including uterine, breast and colon cancer (5).

Epithelial cell dysfunction and senescence has emerged as a central component of the IPF pathophysiology (11,12). The alveolar epithelium consists of alveolar epithelial type 1 (AT1) and alveolar epithelial type 2 cells (AT2). The alveolar surface is mostly covered by AT1 cells, whose thin squamous morphology and intimate contact with the adjacent capillary plexus permit efficient gas exchange (13). Although loss of AT1 cells is considered to be a cardinal feature of the IPF histology, accumulating evidence has revealed AT2 cells to also serve an important role in IPF (14,15). This is in part due to its function in alveolar niche homeostasis through the production of pulmonary surfactants and as a progenitor cell for both self-renewal and transdifferentiation into AT1 cells if needed (13). In particular, single-cell RNA sequencing has previously identified a cell population expressing both AT2 (SOX4, SOX9, COL1A1 and FN1) and AT1 (COL1A1, FN1 and SCGB1A1) markers in IPF lungs, suggesting a subset of epithelial cells transitioning between the AT1 and AT2 phenotype (16).

In eukaryotic cells, a substantial proportion of signal transduction activity is facilitated by protein kinases, which is achieved through phosphorylation of target substrates (17,18). This process serves pivotal roles in the regulation of a wide variety of cellular functions, including proliferation, differentiation, metabolism and programmed cell death (19,20). In humans, protein kinases can be categorized into nine groups based on the evolutionary relationships of their catalytic domains (17,18,21). One such group is one consisting of cAMP-dependent protein kinase A, cGMP-dependent protein kinase G and phospholipid-dependent protein kinase C (AGC), collectively known as AGC kinases (21).

AGC kinases form a highly conserved group of kinases that are ubiquitously distributed across different orders of eukaryotic organisms (21). Members of the AGC kinases group have been reported to regulate different cellular processes, where their targets may have therapeutic implications for various human diseases, including but not limited to cancer, diabetes, obesity, immunological disorders, inflammation, neurological disorders, viral infections and muscular dystrophies (21-24). Therefore, targeting members of the AGC kinases may prove to be a potential method of treatment for PF.

The present review summarizes the reported significant effects of AGC kinases on the pathological procession of PF, before discussing their potential as molecular targets for the treatment of this disease. In addition, focus will be placed on the role of different families of AGC kinases in PF.

2. AGC kinases

The AGC kinase group is comprised of 63 serine/threonine protein kinases that are evolutionarily related. This group includes the protein kinase G and protein kinase C families of kinases, Akt/protein kinase B, Aurora kinases, ribosomal protein S6 kinases and the phosphoinositide-dependent kinases (17,22,24). In addition, the majority of AGC kinases each have multiple isoforms and splice variants, increasing the complexity of this family of kinases (25).

AGC kinases typically exhibit a conserved fold that is characterized by a catalytic domain consisting of a small N-terminal lobe and a large C-terminal lobe (21). The predominant secondary structure of the large C-terminal lobe is α-helical, whereas the small N-terminal lobe is comprised of a single helix (α-C) and a 5-stranded β-sheet (22). An ATP-binding site is located between the two lobes (21,26), where the bound ATP serves as the phosphate donor during phosphorylation (25). The activation loop originating from an Aspartate-Phenylalanine-Glycine motif is also situated amidst the large and the small lobe (21). In addition. the majority of AGC kinases contain a conserved catalytic core with a C-terminal hydrophobic motif (HM) sequence (21). This HM sequence is known to bind to a co-evolved hydrophobic site in the small lobe of the catalytic core, which is referred to as the 3-phosphoinositide-dependent protein kinase-1-interacting fragment (PIF)-pocket (Fig. 2) (27,28). According to a previous study, the PIF-pocket is proposed to be a central and common on-off switch in the AGC kinases (22). Apart from the conserved catalytic domain, the AGC kinases group contains various functional domains. The AGC kinases can be classified into 14 families and 21 subfamilies based on homology outside the catalytic domain (17,25).

AGC kinases serve a crucial role in regulating a multitude of cellular functions, including but not limited to cell cycle progression, cellular differentiation, cell survival and apoptosis (21). In both animals and yeast, AGC kinases have been documented to serve as key mediators that are capable of transducing signaling cascades initiated by secondary messengers through substrate phosphorylation (29,30). In plants, AGC kinases have been demonstrated to serve indispensable roles in diverse cellular and developmental processes including growth, immunity, cell death and defense responses (31-33).

3. AGC kinases in PF

Pyruvate dehydrogenase kinase 1 (PDK1)

PDK1 is a serine/threonine kinase that was initially discovered in previous studies on insulin-activated Akt signaling in the presence of phosphatidylinositol-3,4,5-triphosphate (PIP3) (34-36). PDK1 is a conserved protein kinase that is expressed in eukaryotes (37). PDK1 is mainly located in the cytoplasm, but under certain conditions it can be induced to translocate into the nucleus (38). PDK1 was originally considered to be a regulator of glycolysis in the cytoplasm (39-41). Subsequent studies have revealed that PDK1 can regulate a number of physiological processes, such as blood vessel formation, metabolism and development (42,43). In addition, the pathological processes of Alzheimer's disease (44), diabetes (45) and cancer (46,47) have all been reported to be caused at least in part by PDK1 activity (39).

Previous studies have revealed that PDK1 serves a role in the regulation of PF. The PDK1 gene was previously shown to be a direct target gene of hypoxia-inducible factor-1 (HIF-1) (48,49). Glycolytic metabolism, which is mediated by PDK-1, serves a crucial role in the progression of PF (50-52). Goodwin et al (53) previously reported that hypoxia markedly enhanced transforming growth factor-β (TGF-β)-induced myofibroblast differentiation in fibrotic lesions via HIF-1α. However, overexpression of PDK1 was sufficient in activating glycolysis and potentiate myofibroblast differentiation regardless of the existence of HIF-1α. Additionally, bleomycin (BLM)-induced PF can be significantly attenuated by using dichloroacetate, a potent PDK inhibitor (54,55). Yang et al (56) revealed that PDK1 knockdown can attenuate PF by inhibiting the NF-κB/p65 signaling pathway. Mannan-binding lectin (MBL) can interact with and ubiquitinate PDK1 to inhibit epithelial-mesenchymal transition (EMT) in PF by attenuating store-operated calcium entry (SOCE) signaling (57). However, the specific mechanism of PDK1 in IPF remains unclear.

Rho-associated coiled-coil-forming protein kinase (ROCK)

ROCK is a downstream target protein of Rho and has been implicated in a wide range of cell functions, such as proliferation, migration, adhesion, apoptosis and differentiation (58-60). ROCK has two isoforms, namely ROCK-I and ROCK-II, which regulate cytoskeletal reorganization by phosphorylating myosin phosphatase to increase the phosphorylation level of myosin light chain (61). In addition to brain and muscle tissues, the expression of ROCK-I is widespread, whilst ROCK-II expression tends to be limited to the brain and muscle, especially in the smooth muscle (62). However, the functional differences between ROCK-I and ROCK-II remain unclear (59).

ROCK-II mRNA expression has been previously revealed to be increased in a murine model of lung fibrosis induced by BLM (59). The Rho/ROCK signaling pathway can be inhibited to prevent fibrosis by decreasing the levels of inflammatory cells (macrophages, neutrophils and lymphocytes) and cytokine (TGF-β1, connective tissue growth factor (CTGF) and plasminogen activator inhibitor (PAI)-1 levels (59,63). Shimizu et al (64) demonstrated that the expression and activity of ROCK-II was increased in several types of lung cells in patients with IPF, including bronchial epithelial cells, airway smooth muscle cells, vascular smooth muscle cells and fibroblasts (64). The RhoA/ROCK-I signaling pathway has also been demonstrated to promote the migration of lung fibroblasts and synthesis of collagen by myofibroblasts, both of which can exacerbate PF (65). Rho/ROCK inhibitors, such as Fasudil, have been shown to attenuate BLM-induced lung fibrosis by suppressing the recruitment of inflammatory cells such as neutrophils and reducing the production of TGF-β1, CTGF, α-smooth muscle actin (α-SMA) and PAI-1 in BLM-induced mouse lungs (63). Recently, compound 9b, a novel selective inhibitor of ROCK-II, has demonstrated marked anti-PF effects by suppressing the expression of α-SMA and collagen I in BLM-induced IPF mice model (66). Notably, dual pharmacological inhibition of ROCK-I and -II was found to counteract TGF-β-induced PF in an organoid assay, which included freshly isolated EpCAM+ mouse lung cells co-cultured with human lung fibroblasts (67). However, it should be emphasized that although the main role of ROCK in PF has been established, the precise regulatory mechanisms mediated by Rho/ROCK signaling require further clarification.

Large tumor suppressor 1/2 (LATS1/2)

LATS1 and 2 are important components of the kinase cascades in the Hippo signal pathway in mammalian cells (68-70). A number of studies have demonstrated that LATS2 and its downstream signaling pathway have a vital impact on the proliferation, migration, differentiation and immunomodulation of mesenchymal stem cells (MSCs) (71,72). Dong and Li (71) previously revealed that LATS2-underexpressing bone marrow-derived MSCs (transfected with LATS2-interfering lentivirus vector) can repair the alveolar epithelium damaged by lipopolysaccharide in a mouse model of acute lung injury (ALI).

Lung injury has been reported to trigger the fibrotic process (73). Previous studies have demonstrated that MSCs can reduce collagen fiber deposition and alleviate early-stage PF in mice with ALI (74,75). Dong and Li (71) revealed that this effect is amplified in bone marrow-derived mesenchymal stem cells with low expression levels of LATS2 (due to transfection with LATS2-interfering lentivirus vector). However, further studies are required to investigate the specific mechanisms of LATS in PF.

AKT

AKT, also known as PKB, has three isoforms in mammals, namely AKT1, AKT2 and AKT3. It can regulate numerous cellular processes, such as cell survival, proliferation, differentiation and intermediary metabolism (76-81). Specifically, it has been previously revealed that both AKT1 and AKT2 can modulate the migration and invasion of cancer cells. AKT1 can stimulate prostate cancer cell motility, whereas AKT2 inhibits motility and migration in breast cancer and ovarian cancer cells (82,83). Since AKT3 is primarily expressed in the brain tissue and has only been reported to serve a role in neuronal development (84), research on the role of AKT in PF has mainly concentrated on AKT1 and AKT2(81).

Previous studies have demonstrated that TGF-β1 can regulate the activation of AKT in myofibroblasts and that inhibiting the function of AKT can alleviate TGF-β1-induced PF (85,86). It has been found that AKT1 and AKT2 can mediate significant roles in regulating the function of alveolar macrophages in IPF (87,88). Specifically, AKT1 can promote macrophage mitochondrial reactive oxygen species (ROS) and mitophagy, as well as increase TGF-β1 expression, resulting in the development of fibrosis (88). In addition, the pro-fibrotic cytokine IL-13, can be upregulated by AKT1 in macrophages in PF (89). Nie et al (87) revealed that AKT2 phosphorylation is upregulated in the tissues of patients with PF. AKT2 deficiency protects against BLM-induced PF and inflammation (87). In conclusion, AKT may serve an important role in the development of IPF, suggesting that it can be a potential molecular target for its therapeutic intervention.

Protein kinase C (PKC)

PKC is a type of phospholipid-dependent serine/threonine kinase for which 12 isozymes have been identified (90). PKCs are classified into three subfamilies, based on structural and activation characteristics: conventional or classic PKCs (cPKCs: α, βI, βII and γ), novel or non-classic PKCs (nPKCs: δ, ε, η and θ), and atypical PKCs (aPKC: ζ, ι and λ) (91). PKC isozymes participate in signal transduction by either directly or indirectly activating or inactivating target proteins through phosphorylation (92). PKC has been documented to mediate various cellular processes, including proliferation, migration, apoptosis, adhesion and differentiation (93-96).

The role of PKC-δ in IPF remains controversial, despite its reported involvement in the progression of PF (97). PKC-δ has been reported to inhibit NF-κB signaling by enhancing the activity and stability of A20, which is an endogenous negative regulator of NF-κB (97). In addition, the deficiency of PKC-δ has been reported to increase the expression of proinflammatory cytokines to exacerbate inflammation and PF induced by BLM (97), suggesting that PKC-δ may serve a protective role in IPF. Previous studies have demonstrated that the inhibitor of PKC-δ rottlerin can downregulate the expression of type I and type III collagen gene, and suppress the type I collagen production in cultured dermal fibroblasts derived from patients with systemic sclerosis (98). Additionally, Song et al (99) revealed that thrombin induces EMT and collagen I secretion by activating protease-activated receptor (PAR-1), PKC (α/β, δ and ε) and ERK1/2 in A549 cells. A549 is an adenocarcinomic human alveolar basal epithelial cell line, that is widely used as a model of alveolar epithelial-like behavior in IPF study (100). Therefore, targeting PAR-1 or specific PKC isoforms (α, β, δ and ε) may halt the fibrotic process in human IPF by preventing thrombin-induced EMT. Results reported by the aforementioned studies suggest that PKC-δ may promote IPF. However, the possible link between PKC and IPF require further studies.

Ribosome protein S6 kinase (RPS6K)

RPS6Ks can be divided into two subfamilies, p90 ribosomal S6 kinase (RSK) and p70 ribosomal S6 kinase (p70S6K). The p70S6K subfamily has two members of the p70S6K (S6K1 and S6K2), whilst the RSK subfamily has four members (RSK1-4) (101,102). RSK is activated by the ERK signaling pathway. p70S6K is activated through a complex network of signaling molecules, and mTOR serine/threonine kinase is necessary for its full activation (103). These kinases are involved in various signaling pathways and can regulate multiple cellular processes, such as cell proliferation, differentiation, growth, transformation and apoptosis (104-108).

Madala et al (109) previously revealed an increase in S6 phosphorylation in the airway and alveolar epithelium and in the mesenchyme of advanced subpleural fibrotic regions of TGF-α-induced PF mice. The specific targeted inhibition of the S6K with the small molecule inhibitor LY-2584702 attenuates TGF-α and platelet-derived growth factor-β-induced proliferation of pulmonary fibroblasts (109). In another study, Han et al (110) revealed that rapamycin, an mTOR inhibitor, can attenuate BLM-induced PF and EMT by decreasing S6K- and TGF-β1-induced Smad2/3 phosphorylation. In addition, S6K was also found to enhance proliferation and fibroblast-to-myofibroblast transition in human embryonic lung fibroblasts (111). Kim et al (112) revealed that inhibition of RSK suppressed TGF-β1-induced ECM accumulation and EMT in lung epithelial cells and fibroblasts. These findings suggest that RPS6K may also have a role in the development of IPF. However, the specific mechanism underlying IPF progression requires further elucidation.

4. Conclusions

The AGC kinases form a widely conserved family of protein kinases that have been implicated in various pathologies, including cancer, metabolic disorders, cardiovascular disease, immunological disorders and neurological disorders. Accumulating evidence has shown that AGC kinases can exert important roles in IPF through distinct mechanistic pathways (Table I). Several inhibitors of AGC kinases such as RSK-inhibitor peptide (112), dichloroacetate (53), BX795 and BX912(113) have been found to attenuate PF. However, additional research is required to fully comprehend the contribution of AGC kinases towards IPF. Identification of AGC kinases with the potential to serve as therapeutic targets of IPF may facilitate the discovery of novel drugs for IPF treatment.

Table I

Mechanisms of AGC kinases in IPF.

Table I

Mechanisms of AGC kinases in IPF.

First author(s), yearAGC kinasesIsoformsMechanisms(Refs.)
Yang et al, 2021PDK1 NF-κB/p65 pathway(56)
Goodwin et al, 2018  Hypoxia-inducible factor-1a/3-phosphoinositide-dependent protein kinase-1-mediated glycolytic reprogramming(53)
Jiang et al, 2012ROCK Rho/ROCK signaling pathway(63)
Shimizu et al, 2014  Rho/ROCK signaling pathway(64)
Dong and Li, 2019Large tumor suppressor Hippo signaling pathway(71)
Wang et al, 2022AKT PI3K/AKT signaling pathway(81)
Nie et al, 2019 AKT1Modulating IL-13 expression in macrophages(89)
Nie et al, 2017 AKT2AKT2/Forkhead box O3a signaling pathway(87)
Song et al, 2013PKC Protease-activated receptor-1/PKC/ERK pathways(99)
Wang et al, 2020 PKC-δNF-κB/A20 signaling(97)
Kim et al, 2020p90 ribosomal S6 kinase TGF-β1/smad3 signaling(112)
Madala et al, 2016S6K S6K signaling(109)

[i] ROCK, Rho-associated coiled-coil-forming protein kinase; PKC, protein kinase C; S6K, p70 ribosomal S6 kinase.

At present, to the best of our knowledge, only a small number of AGC kinases have been found to be involved in regulating the pathological process of PF. To further explore the key role of AGC kinases comprehensively in the pathogenesis of PF, more functions of AGC kinases in PF need to be explored. As single-cell sequencing and spatial proteomics technology advance, the distinct functions mediated by AGC kinase in various cell types during different stages of PF will be elucidated (114-116). In addition, continuous advancements in organoid technology are expected to facilitate studies into the microenvironment of lung tissues in the pathological process of PF in the future, where the role of AGC kinases should also be investigated. Understanding the specific substrates and associated signaling pathways by AGC kinases in the regulation of PF will also be a focus of future attention. Based on the results and findings of existing studies, it would be of benefit to screen small molecule inhibitors or targeted drugs for AGC kinase and conduct relevant clinical trials, providing more effective treatment options and strategies for patients with IPF.

Acknowledgements

Not applicable.

Funding

Funding: The present study was supported by grants from the National Natural Science Foundation of China (grant nos. 82060023 and 82160133) and the Jiangxi Provincial Natural Science Foundation (grant nos. 20202ACBL206015, 20224BAB206007 and 20212ACB216005).

Availability of data and materials

Not applicable.

Authors' contribution

YL conceived the study and revised the manuscript. CM prepared the figures and wrote the manuscript. TC, XH and CX participated in the writing of the manuscript. SC reviewed the manuscript. All authors have read and approved the final manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Noble PW, Barkauskas CE and Jiang D: Pulmonary fibrosis: Patterns and perpetrators. J Clin Invest. 122:2756–2762. 2012.PubMed/NCBI View Article : Google Scholar

2 

Kreuter M, Ladner UM, Costabel U, Jonigk D and Heussel CP: The diagnosis and treatment of pulmonary fibrosis. Dtsch Arztebl Int. 118:152–162. 2021.PubMed/NCBI View Article : Google Scholar

3 

Richeldi L, Collard HR and Jones MG: Idiopathic pulmonary fibrosis. Lancet. 389:1941–1952. 2017.PubMed/NCBI View Article : Google Scholar

4 

Günther A, Korfei M, Mahavadi P, von der Beck D, Ruppert C and Markart P: Unravelling the progressive pathophysiology of idiopathic pulmonary fibrosis. Eur Respir Rev. 21:152–160. 2012.PubMed/NCBI View Article : Google Scholar

5 

Raghu G, Collard HR, Egan JJ, Martinez FJ, Behr J, Brown KK, Colby TV, Cordier JF, Flaherty KR, Lasky JA, et al: An official ATS/ERS/JRS/ALAT statement: Idiopathic pulmonary fibrosis: Evidence-based guidelines for diagnosis and management. Am J Respir Crit Care Med. 183:788–824. 2011.PubMed/NCBI View Article : Google Scholar

6 

Lederer DJ and Martinez FJ: Idiopathic pulmonary fibrosis. N Engl J Med. 378:1811–1823. 2018.PubMed/NCBI View Article : Google Scholar

7 

Abuserewa ST, Duff R and Becker G: Treatment of idiopathic pulmonary fibrosis. Cureus. 13(e15360)2021.PubMed/NCBI View Article : Google Scholar

8 

Khor YH: Antifibrotic therapy for idiopathic pulmonary fibrosis: Combining real world and clinical trials for totality of evidence. Chest. 160:1589–1591. 2021.PubMed/NCBI View Article : Google Scholar

9 

Desai O, Winkler J, Minasyan M and Herzog EL: The role of immune and inflammatory cells in idiopathic pulmonary fibrosis. Front Med (Lausanne). 5(43)2018.PubMed/NCBI View Article : Google Scholar

10 

Fujimoto H, Kobayashi T and Azuma A: Idiopathic pulmonary fibrosis: Treatment and prognosis. Clin Med Insights Circ Respir Pulm Med. 9 (Suppl 1):S179–S185. 2016.PubMed/NCBI View Article : Google Scholar

11 

Selman M and Pardo A: The leading role of epithelial cells in the pathogenesis of idiopathic pulmonary fibrosis. Cell Signal. 66(109482)2020.PubMed/NCBI View Article : Google Scholar

12 

Tu M, Wei T, Jia Y, Wang Y and Wu J: Molecular mechanisms of alveolar epithelial cell senescence and idiopathic pulmonary fibrosis: A narrative review. J Thorac Dis. 15:186–203. 2023.PubMed/NCBI View Article : Google Scholar

13 

Katzen J and Beers MF: Contributions of alveolar epithelial cell quality control to pulmonary fibrosis. J Clin Invest. 130:5088–5099. 2020.PubMed/NCBI View Article : Google Scholar

14 

Parimon T, Yao C, Stripp BR, Noble PW and Chen P: Alveolar epithelial type II cells as drivers of lung fibrosis in idiopathic pulmonary fibrosis. Int J Mol Sci. 21(2269)2020.PubMed/NCBI View Article : Google Scholar

15 

Zhu W, Tan C and Zhang J: Alveolar epithelial type 2 cell dysfunction in idiopathic pulmonary fibrosis. Lung. 200:539–547. 2022.PubMed/NCBI View Article : Google Scholar

16 

Habermann AC, Gutierrez AJ, Bui LT, Yahn SL, Winters NI, Calvi CL, Peter L, Chung MI, Taylor CJ, Jetter C, et al: Single-cell RNA sequencing reveals profibrotic roles of distinct epithelial and mesenchymal lineages in pulmonary fibrosis. Sci Adv. 6(eaba1972)2020.PubMed/NCBI View Article : Google Scholar

17 

Manning G, Whyte DB, Martinez R, Hunter T and Sudarsanam S: The protein kinase complement of the human genome. Science. 298:1912–1934. 2002.PubMed/NCBI View Article : Google Scholar

18 

Roskoski R Jr: A historical overview of protein kinases and their targeted small molecule inhibitors. Pharmacol Res. 100:1–23. 2015.PubMed/NCBI View Article : Google Scholar

19 

Deribe YL, Pawson T and Dikic I: Post-translational modifications in signal integration. Nat Struct Mol Biol. 17:666–672. 2010.PubMed/NCBI View Article : Google Scholar

20 

Attwood MM, Fabbro D, Sokolov AV, Knapp S and Schiöth HB: Trends in kinase drug discovery: Targets, indications and inhibitor design. Nat Rev Drug Discov. 20:839–861. 2021.PubMed/NCBI View Article : Google Scholar

21 

Arencibia JM, Pastor-Flores D, Bauer AF, Schulze JO and Biondi RM: AGC protein kinases: From structural mechanism of regulation to allosteric drug development for the treatment of human diseases. Biochim Biophys Acta. 1834:1302–1321. 2013.PubMed/NCBI View Article : Google Scholar

22 

Leroux AE, Schulze JO and Biondi RM: AGC kinases, mechanisms of regulation and innovative drug development. Semin Cancer Biol. 48:1–17. 2018.PubMed/NCBI View Article : Google Scholar

23 

Rath N and Olson MF: Rho-associated kinases in tumorigenesis: Re-considering ROCK inhibition for cancer therapy. EMBO Rep. 13:900–908. 2012.PubMed/NCBI View Article : Google Scholar

24 

Turnham RE and Scott JD: Protein kinase A catalytic subunit isoform PRKACA; history, function and physiology. Gene. 577:101–108. 2016.PubMed/NCBI View Article : Google Scholar

25 

Pearce LR, Komander D and Alessi DR: The nuts and bolts of AGC protein kinases. Nat Rev Mol Cell Biol. 11:9–22. 2010.PubMed/NCBI View Article : Google Scholar

26 

Zheng J, Knighton DR, ten Eyck LF, Karlsson R, Xuong N, Taylor SS and Sowadski JM: Crystal structure of the catalytic subunit of cAMP-dependent protein kinase complexed with MgATP and peptide inhibitor. Biochemistry. 32:2154–2161. 1993.PubMed/NCBI View Article : Google Scholar

27 

Biondi RM, Cheung PC, Casamayor A, Deak M, Currie RA and Alessi DR: Identification of a pocket in the PDK1 kinase domain that interacts with PIF and the C-terminal residues of PKA. EMBO J. 19:979–988. 2000.PubMed/NCBI View Article : Google Scholar

28 

Biondi RM, Komander D, Thomas CC, Lizcano JM, Deak M, Alessi DR and van Aalten DM: High resolution crystal structure of the human PDK1 catalytic domain defines the regulatory phosphopeptide docking site. EMBO J. 21:4219–4228. 2002.PubMed/NCBI View Article : Google Scholar

29 

Zhang Y and McCormick S: AGCVIII kinases: At the crossroads of cellular signaling. Trends Plant Sci. 14:689–695. 2009.PubMed/NCBI View Article : Google Scholar

30 

Sobko A: Systems biology of AGC kinases in fungi. Sci STKE. 2006(re9)2006.PubMed/NCBI View Article : Google Scholar

31 

Lanassa Bassukas AE, Xiao Y and Schwechheimer C: Phosphorylation control of PIN auxin transporters. Curr Opin Plant Biol. 65(102146)2022.PubMed/NCBI View Article : Google Scholar

32 

Jiang Y, Liu X, Zhou M, Yang J, Ke S and Li Y: Genome-wide identification of the AGC protein kinase gene family related to photosynthesis in rice (Oryza sativa). Int J Mol Sci. 23(12557)2022.PubMed/NCBI View Article : Google Scholar

33 

Glanc M, Van Gelderen K, Hoermayer L, Tan S, Naramoto S, Zhang X, Domjan D, Včelařová L, Hauschild R, Johnson A, et al: AGC kinases and MAB4/MEL proteins maintain PIN polarity by limiting lateral diffusion in plant cells. Curr Biol. 31:1918–1930.e5. 2021.PubMed/NCBI View Article : Google Scholar

34 

Wick KL and Liu F: A new molecular target of insulin action: Regulating the pivotal PDK1. Curr Drug Targets Immune Endocr Metabol Disord. 1:209–221. 2001.PubMed/NCBI View Article : Google Scholar

35 

Alessi DR, James SR, Downes CP, Holmes AB, Gaffney PR, Reese CB and Cohen P: Characterization of a 3-phosphoinositide-dependent protein kinase which phosphorylates and activates protein kinase Balpha. Curr Biol. 7:261–269. 1997.PubMed/NCBI View Article : Google Scholar

36 

Stokoe D, Stephens LR, Copeland T, Gaffney PR, Reese CB, Painter GF, Holmes AB, McCormick F and Hawkins PT: Dual role of phosphatidylinositol-3,4,5-trisphosphate in the activation of protein kinase B. Science. 277:567–570. 1997.PubMed/NCBI View Article : Google Scholar

37 

Dittrich ACN and Devarenne TP: Perspectives in PDK1 evolution: Insights from photosynthetic and non-photosynthetic organisms. Plant Signal Behav. 7:642–649. 2012.PubMed/NCBI View Article : Google Scholar

38 

Scheid MP, Parsons M and Woodgett JR: Phosphoinositide-dependent phosphorylation of PDK1 regulates nuclear translocation. Mol Cell Biol. 25:2347–2363. 2005.PubMed/NCBI View Article : Google Scholar

39 

Gagliardi PA, di Blasio L and Primo L: PDK1: A signaling hub for cell migration and tumor invasion. Biochim Biophys Acta. 1856:178–188. 2015.PubMed/NCBI View Article : Google Scholar

40 

Cohen P, Alessi DR and Cross DA: PDK1, one of the missing links in insulin signal transduction? FEBS Lett. 410:3–10. 1997.PubMed/NCBI View Article : Google Scholar

41 

Zhou Y, Guo Y, Ran M, Shan W, Granchi C, Giovannetti E, Minutolo F, Peters GJ and Tam KY: Combined inhibition of pyruvate dehydrogenase kinase 1 and lactate dehydrogenase a induces metabolic and signaling reprogramming and enhances lung adenocarcinoma cell killing. Cancer Lett. 577(216425)2023.PubMed/NCBI View Article : Google Scholar

42 

Feng Q, Di R, Tao F, Chang Z, Lu S, Fan W, Shan C, Li X and Yang Z: PDK1 regulates vascular remodeling and promotes epithelial-mesenchymal transition in cardiac development. Mol Cell Biol. 30:3711–3721. 2010.PubMed/NCBI View Article : Google Scholar

43 

Lawlor MA, Mora A, Ashby PR, Williams MR, Murray-Tait V, Malone L, Prescott AR, Lucocq JM and Alessi DR: Essential role of PDK1 in regulating cell size and development in mice. EMBO J. 21:3728–3738. 2002.PubMed/NCBI View Article : Google Scholar

44 

Pietri M, Dakowski C, Hannaoui S, Alleaume-Butaux A, Hernandez-Rapp J, Ragagnin A, Mouillet-Richard S, Haik S, Bailly Y, Peyrin JM, et al: PDK1 decreases TACE-mediated α-secretase activity and promotes disease progression in prion and Alzheimer's diseases. Nat Med. 19:1124–1131. 2013.PubMed/NCBI View Article : Google Scholar

45 

Hashimoto N, Kido Y, Uchida T, Asahara S, Shigeyama Y, Matsuda T, Takeda A, Tsuchihashi D, Nishizawa A, Ogawa W, et al: Ablation of PDK1 in pancreatic beta cells induces diabetes as a result of loss of beta cell mass. Nat Genet. 38:589–593. 2006.PubMed/NCBI View Article : Google Scholar

46 

Choucair KA, Guérard KP, Ejdelman J, Chevalier S, Yoshimoto M, Scarlata E, Fazli L, Sircar K, Squire JA, Brimo F, et al: The 16p13.3 (PDPK1) genomic gain in prostate cancer: A potential role in disease progression. Transl Oncol. 5:453–460. 2012.PubMed/NCBI View Article : Google Scholar

47 

Maurer M, Su T, Saal LH, Koujak S, Hopkins BD, Barkley CR, Wu J, Nandula S, Dutta B, Xie Y, et al: 3-Phosphoinositide-dependent kinase 1 potentiates upstream lesions on the phosphatidylinositol 3-kinase pathway in breast carcinoma. Cancer Res. 69:6299–6306. 2009.PubMed/NCBI View Article : Google Scholar

48 

Kim JW, Tchernyshyov I, Semenza GL and Dang CV: HIF-1-mediated expression of pyruvate dehydrogenase kinase: A metabolic switch required for cellular adaptation to hypoxia. Cell Metab. 3:177–185. 2006.PubMed/NCBI View Article : Google Scholar

49 

Papandreou I, Cairns RA, Fontana L, Lim AL and Denko NC: HIF-1 mediates adaptation to hypoxia by actively downregulating mitochondrial oxygen consumption. Cell Metab. 3:187–197. 2006.PubMed/NCBI View Article : Google Scholar

50 

Li J, Zhai X, Sun X, Cao S, Yuan Q and Wang J: Metabolic reprogramming of pulmonary fibrosis. Front Pharmacol. 13(1031890)2022.PubMed/NCBI View Article : Google Scholar

51 

Hamanaka RB and Mutlu GM: Metabolic requirements of pulmonary fibrosis: Role of fibroblast metabolism. FEBS J. 288:6331–6352. 2021.PubMed/NCBI View Article : Google Scholar

52 

Henderson J and O'Reilly S: The emerging role of metabolism in fibrosis. Trends Endocrinol Metab. 32:639–653. 2021.PubMed/NCBI View Article : Google Scholar

53 

Goodwin J, Choi H, Hsieh MH, Neugent ML, Ahn JM, Hayenga HN, Singh PK, Shackelford DB, Lee IK, Shulaev V, et al: Targeting hypoxia-inducible factor-1α/pyruvate dehydrogenase kinase 1 axis by dichloroacetate suppresses bleomycin-induced pulmonary fibrosis. Am J Respir Cell Mol Biol. 58:216–231. 2018.PubMed/NCBI View Article : Google Scholar

54 

Stacpoole PW: Review of the pharmacologic and therapeutic effects of diisopropylammonium dichloroacetate (DIPA). J Clin Pharmacol J New Drugs. 9:282–291. 1969.PubMed/NCBI

55 

Stacpoole PW, Kurtz TL, Han Z and Langaee T: Role of dichloroacetate in the treatment of genetic mitochondrial diseases. Adv Drug Deliv Rev. 60:1478–1487. 2008.PubMed/NCBI View Article : Google Scholar

56 

Yang K, Li B and Chen J: Knockdown of phosphoinositide-dependent kinase 1 (PDK1) inhibits fibrosis and inflammation in lipopolysaccharide-induced acute lung injury rat model by attenuating NF-κB/p65 pathway activation. Ann Transl Med. 9(1671)2021.PubMed/NCBI View Article : Google Scholar

57 

Liu Y, Xie X, Wang P, Luo J, Chen Y, Xu Q, Zhou J, Lu X, Zhao J, Chen Z and Zuo D: Mannan-binding lectin reduces epithelial-mesenchymal transition in pulmonary fibrosis via inactivating the store-operated calcium entry machinery. J Innate Immun. 15:37–49. 2023.PubMed/NCBI View Article : Google Scholar

58 

Loirand G, Guérin P and Pacaud P: Rho kinases in cardiovascular physiology and pathophysiology. Circ Res. 98:322–334. 2006.PubMed/NCBI View Article : Google Scholar

59 

Shimizu Y, Dobashi K, Iizuka K, Horie T, Suzuki K, Tukagoshi H, Nakazawa T, Nakazato Y and Mori M: Contribution of small GTPase Rho and its target protein rock in a murine model of lung fibrosis. Am J Respir Crit Care Med. 163:210–217. 2001.PubMed/NCBI View Article : Google Scholar

60 

Barcelo J, Samain R and Sanz-Moreno V: Preclinical to clinical utility of ROCK inhibitors in cancer. Trends Cancer. 9:250–263. 2023.PubMed/NCBI View Article : Google Scholar

61 

Shimokawa H and Takeshita A: Rho-kinase is an important therapeutic target in cardiovascular medicine. Arterioscler Thromb Vasc Biol. 25:1767–1775. 2005.PubMed/NCBI View Article : Google Scholar

62 

Knipe RS, Tager AM and Liao JK: The Rho kinases: Critical mediators of multiple profibrotic processes and rational targets for new therapies for pulmonary fibrosis. Pharmacol Rev. 67:103–117. 2015.PubMed/NCBI View Article : Google Scholar

63 

Jiang C, Huang H, Liu J, Wang Y, Lu Z and Xu Z: Fasudil, a Rho-kinase inhibitor, attenuates bleomycin-induced pulmonary fibrosis in mice. Int J Mol Sci. 13:8293–8307. 2012.PubMed/NCBI View Article : Google Scholar

64 

Shimizu Y, Dobashi K, Sano T and Yamada M: ROCK activation in lung of idiopathic pulmonary fibrosis with oxidative stress. Int J Immunopathol Pharmacol. 27:37–44. 2014.PubMed/NCBI View Article : Google Scholar

65 

Ghatak S, Hascall VC, Markwald RR, Feghali-Bostwick C, Artlett CM, Gooz M, Bogatkevich GS, Atanelishvili I, Silver RM, Wood J, et al: Transforming growth factor β1 (TGFβ1)-induced CD44V6-NOX4 signaling in pathogenesis of idiopathic pulmonary fibrosis. J Biol Chem. 292:10490–10519. 2017.PubMed/NCBI View Article : Google Scholar

66 

Fu S, Wen Y, Peng B, Tang M, Shi M, Liu J, Yang Y, Si W, Guo Y, Li X, et al: Discovery of indoline-based derivatives as effective ROCK2 inhibitors for the potential new treatment of idiopathic pulmonary fibrosis. Bioorg Chem. 137(106539)2023.PubMed/NCBI View Article : Google Scholar

67 

Wu X, Verschut V, Woest ME, Ng-Blichfeldt JP, Matias A, Villetti G, Accetta A, Facchinetti F, Gosens R and Kistemaker LEM: Rho-kinase 1/2 inhibition prevents transforming growth factor-β-induced effects on pulmonary remodeling and repair. Front Pharmacol. 11(609509)2021.PubMed/NCBI View Article : Google Scholar

68 

Hong AW, Meng Z and Guan KL: The Hippo pathway in intestinal regeneration and disease. Nat Rev Gastroenterol Hepatol. 13:324–337. 2016.PubMed/NCBI View Article : Google Scholar

69 

Wu Z and Guan KL: Hippo signaling in embryogenesis and development. Trends Biochem Sci. 46:51–63. 2021.PubMed/NCBI View Article : Google Scholar

70 

Landry NM, Rattan SG, Filomeno KL, Meier TW, Meier SC, Foran SJ, Meier CF, Koleini N, Fandrich RR, Kardami E, et al: SKI activates the Hippo pathway via LIMD1 to inhibit cardiac fibroblast activation. Basic Res Cardiol. 116(25)2021.PubMed/NCBI View Article : Google Scholar

71 

Dong L and Li L: Lats2-underexpressing bone marrow-derived mesenchymal stem cells ameliorate LPS-induced acute lung injury in mice. Mediators Inflamm. 2019(4851431)2019.PubMed/NCBI View Article : Google Scholar

72 

Antebi B, Walker KP III, Mohammadipoor A, Rodriguez LA, Montgomery RK, Batchinsky AI and Cancio LC: The effect of acute respiratory distress syndrome on bone marrow-derived mesenchymal stem cells. Stem Cell Res Ther. 9(251)2018.PubMed/NCBI View Article : Google Scholar

73 

Kuwano K, Miyazaki H, Hagimoto N, Kawasaki M, Fujita M, Kunitake R, Kaneko Y and Hara N: The involvement of Fas-Fas ligand pathway in fibrosing lung diseases. Am J Respir Cell Mol Biol. 20:53–60. 1999.PubMed/NCBI View Article : Google Scholar

74 

Cai SX, Liu AR, Chen S, He HL, Chen QH, Xu JY, Pan C, Yang Y, Guo FM, Huang YZ, et al: The orphan receptor tyrosine kinase ROR2 facilitates MSCs to repair lung injury in ARDS animal model. Cell Transplant. 25:1561–1574. 2016.PubMed/NCBI View Article : Google Scholar

75 

Han J, Lu X, Zou L, Xu X and Qiu H: E-prostanoid 2 receptor overexpression promotes mesenchymal stem cell attenuated lung injury. Hum Gene Ther. 27:621–630. 2016.PubMed/NCBI View Article : Google Scholar

76 

Fernández-Hernando C, Ackah E, Yu J, Suárez Y, Murata T, Iwakiri Y, Prendergast J, Miao RQ, Birnbaum MJ and Sessa WC: Loss of Akt1 leads to severe atherosclerosis and occlusive coronary artery disease. Cell Metab. 6:446–457. 2007.PubMed/NCBI View Article : Google Scholar

77 

Iliopoulos D, Polytarchou C, Hatziapostolou M, Kottakis F, Maroulakou IG, Struhl K and Tsichlis PN: MicroRNAs differentially regulated by Akt isoforms control EMT and stem cell renewal in cancer cells. Sci Signal. 2(ra62)2009.PubMed/NCBI View Article : Google Scholar

78 

Revathidevi S and Munirajan AK: Akt in cancer: Mediator and more. Semin Cancer Biol. 59:80–91. 2019.PubMed/NCBI View Article : Google Scholar

79 

Risso G, Blaustein M, Pozzi B, Mammi P and Srebrow A: Akt/PKB: One kinase, many modifications. Biochem J. 468:203–214. 2015.PubMed/NCBI View Article : Google Scholar

80 

Toker A and Yoeli-Lerner M: Akt signaling and cancer: Surviving but not moving on. Cancer Res. 66:3963–3966. 2006.PubMed/NCBI View Article : Google Scholar

81 

Wang J, Hu K, Cai X, Yang B, He Q, Wang J and Weng Q: Targeting PI3K/AKT signaling for treatment of idiopathic pulmonary fibrosis. Acta Pharm Sin B. 12:18–32. 2022.PubMed/NCBI View Article : Google Scholar

82 

Virtakoivu R, Pellinen T, Rantala JK, Perälä M and Ivaska J: Distinct roles of AKT isoforms in regulating β1-integrin activity, migration, and invasion in prostate cancer. Mol Biol Cell. 23:3357–3369. 2012.PubMed/NCBI View Article : Google Scholar

83 

Arboleda MJ, Lyons JF, Kabbinavar FF, Bray MR, Snow BE, Ayala R, Danino M, Karlan BY and Slamon DJ: Overexpression of AKT2/protein kinase Bbeta leads to up-regulation of beta1 integrins, increased invasion, and metastasis of human breast and ovarian cancer cells. Cancer Res. 63:196–206. 2003.PubMed/NCBI

84 

Baek ST, Copeland B, Yun EJ, Kwon SK, Guemez-Gamboa A, Schaffer AE, Kim S, Kang HC, Song S, Mathern GW and Gleeson JG: An AKT3-FOXG1-reelin network underlies defective migration in human focal malformations of cortical development. Nat Med. 21:1445–1454. 2015.PubMed/NCBI View Article : Google Scholar

85 

Kang HR, Lee CG, Homer RJ and Elias JA: Semaphorin 7A plays a critical role in TGF-beta1-induced pulmonary fibrosis. J Exp Med. 204:1083–1093. 2007.PubMed/NCBI View Article : Google Scholar

86 

Horowitz JC, Rogers DS, Sharma V, Vittal R, White ES, Cui Z and Thannickal VJ: Combinatorial activation of FAK and AKT by transforming growth factor-beta1 confers an anoikis-resistant phenotype to myofibroblasts. Cell Signal. 19:761–771. 2007.PubMed/NCBI View Article : Google Scholar

87 

Nie Y, Sun L, Wu Y, Yang Y, Wang J, He H, Hu Y, Chang Y, Liang Q, Zhu J, et al: AKT2 regulates pulmonary inflammation and fibrosis via modulating macrophage activation. J Immunol. 198:4470–4480. 2017.PubMed/NCBI View Article : Google Scholar

88 

Larson-Casey JL, Deshane JS, Ryan AJ, Thannickal VJ and Carter AB: Macrophage Akt1 kinase-mediated mitophagy modulates apoptosis resistance and pulmonary fibrosis. Immunity. 44:582–596. 2016.PubMed/NCBI View Article : Google Scholar

89 

Nie Y, Hu Y, Yu K, Zhang D, Shi Y, Li Y, Sun L and Qian F: Akt1 regulates pulmonary fibrosis via modulating IL-13 expression in macrophages. Innate Immun. 25:451–461. 2019.PubMed/NCBI View Article : Google Scholar

90 

Kazanietz MG and Cooke M: Protein kinase C signaling ‘in’ and ‘to’ the nucleus: Master kinases in transcriptional regulation. J Biol Chem: 105692, 2024 (Epub ahead of print).

91 

Silnitsky S, Rubin SJS, Zerihun M and Qvit N: An update on protein kinases as therapeutic targets-part I: Protein kinase C activation and its role in cancer and cardiovascular diseases. Int J Mol Sci. 24(17600)2023.PubMed/NCBI View Article : Google Scholar

92 

Kang JH, Toita R, Kim CW and Katayama Y: Protein kinase C (PKC) isozyme-specific substrates and their design. Biotechnol Adv. 30:1662–1672. 2012.PubMed/NCBI View Article : Google Scholar

93 

Abe MK, Kartha S, Karpova AY, Li J, Liu PT, Kuo WL and Hershenson MB: Hydrogen peroxide activates extracellular signal-regulated kinase via protein kinase C, Raf-1, and MEK1. Am J Respir Cell Mol Biol. 18:562–569. 1998.PubMed/NCBI View Article : Google Scholar

94 

Barman SA: Potassium channels modulate canine pulmonary vasoreactivity to protein kinase C activation. Am J Physiol. 277:L558–L565. 1999.PubMed/NCBI View Article : Google Scholar

95 

Das M, Stenmark KR, Ruff LJ and Dempsey EC: Selected isozymes of PKC contribute to augmented growth of fetal and neonatal bovine PA adventitial fibroblasts. Am J Physiol. 273:L1276–L1284. 1997.PubMed/NCBI View Article : Google Scholar

96 

Harrington EO, Löffler J, Nelson PR, Kent KC, Simons M and Ware JA: Enhancement of migration by protein kinase Calpha and inhibition of proliferation and cell cycle progression by protein kinase Cdelta in capillary endothelial cells. J Biol Chem. 272:7390–7397. 1997.PubMed/NCBI View Article : Google Scholar

97 

Wang J, Sun L, Nie Y, Duan S, Zhang T, Wang W, Ye RD, Hou S and Qian F: Protein kinase C δ (PKCδ) attenuates bleomycin induced pulmonary fibrosis via inhibiting NF-κB signaling pathway. Front Physiol. 11(367)2020.PubMed/NCBI View Article : Google Scholar

98 

Jimenez SA, Gaidarova S, Saitta B, Sandorfi N, Herrich DJ, Rosenbloom JC, Kucich U, Abrams WR and Rosenbloom J: Role of protein kinase C-delta in the regulation of collagen gene expression in scleroderma fibroblasts. J Clin Invest. 108:1395–1403. 2001.PubMed/NCBI View Article : Google Scholar

99 

Song JS, Kang CM, Park CK and Yoon HK: Thrombin induces epithelial-mesenchymal transition via PAR-1, PKC, and ERK1/2 pathways in A549 cells. Exp Lung Res. 39:336–348. 2013.PubMed/NCBI View Article : Google Scholar

100 

Barosova H, Meldrum K, Karakocak BB, Balog S, Doak SH, Petri-Fink A, Clift MJD and Rothen-Rutishauser B: Inter-laboratory variability of A549 epithelial cells grown under submerged and air-liquid interface conditions. Toxicol In Vitro. 75(105178)2021.PubMed/NCBI View Article : Google Scholar

101 

McMullen JR, Shioi T, Zhang L, Tarnavski O, Sherwood MC, Dorfman AL, Longnus S, Pende M, Martin KA, Blenis J, et al: Deletion of ribosomal S6 kinases does not attenuate pathological, physiological, or insulin-like growth factor 1 receptor-phosphoinositide 3-kinase-induced cardiac hypertrophy. Mol Cell Biol. 24:6231–6240. 2004.PubMed/NCBI View Article : Google Scholar

102 

Ludwik KA and Lannigan DA: Ribosomal S6 kinase (RSK) modulators: A patent review. Expert Opin Ther Pat. 26:1061–1078. 2016.PubMed/NCBI View Article : Google Scholar

103 

Shima H, Pende M, Chen Y, Fumagalli S, Thomas G and Kozma SC: Disruption of the p70(s6k)/p85(s6k) gene reveals a small mouse phenotype and a new functional S6 kinase. EMBO J. 17:6649–6659. 1998.PubMed/NCBI View Article : Google Scholar

104 

Magnuson B, Ekim B and Fingar DC: Regulation and function of ribosomal protein S6 kinase (S6K) within mTOR signalling networks. Biochem J. 441:1–21. 2012.PubMed/NCBI View Article : Google Scholar

105 

Pullen N, Dennis PB, Andjelkovic M, Dufner A, Kozma SC, Hemmings BA and Thomas G: Phosphorylation and activation of p70s6k by PDK1. Science. 279:707–710. 1998.PubMed/NCBI View Article : Google Scholar

106 

Roux PP, Shahbazian D, Vu H, Holz MK, Cohen MS, Taunton J, Sonenberg N and Blenis J: RAS/ERK signaling promotes site-specific ribosomal protein S6 phosphorylation via RSK and stimulates cap-dependent translation. J Biol Chem. 282:14056–14064. 2007.PubMed/NCBI View Article : Google Scholar

107 

Soares HP, Ni Y, Kisfalvi K, Sinnett-Smith J and Rozengurt E: Different patterns of Akt and ERK feedback activation in response to rapamycin, active-site mTOR inhibitors and metformin in pancreatic cancer cells. PLoS One. 8(e57289)2013.PubMed/NCBI View Article : Google Scholar

108 

Frödin M and Gammeltoft S: Role and regulation of 90 kDa ribosomal S6 kinase (RSK) in signal transduction. Mol Cell Endocrinol. 151:65–77. 1999.PubMed/NCBI View Article : Google Scholar

109 

Madala SK, Thomas G, Edukulla R, Davidson C, Schmidt S, Schehr A and Hardie WD: p70 ribosomal S6 kinase regulates subpleural fibrosis following transforming growth factor-α expression in the lung. Am J Physiol Lung Cell Mol Physiol. 310:L175–L186. 2016.PubMed/NCBI View Article : Google Scholar

110 

Han Q, Lin L, Zhao B, Wang N and Liu X: Inhibition of mTOR ameliorates bleomycin-induced pulmonary fibrosis by regulating epithelial-mesenchymal transition. Biochem Biophys Res Commun. 500:839–845. 2018.PubMed/NCBI View Article : Google Scholar

111 

Zou W, Zhang X, Zhao M, Zhou Q and Hu X: Cellular proliferation and differentiation induced by single-layer molybdenum disulfide and mediation mechanisms of proteins via the Akt-mTOR-p70S6K signaling pathway. Nanotoxicology. 11:781–793. 2017.PubMed/NCBI View Article : Google Scholar

112 

Kim S, Han JH, Kim S, Lee H, Kim JR, Lim JH and Woo CH: p90RSK inhibition ameliorates TGF-β1 signaling and pulmonary fibrosis by inhibiting smad3 transcriptional activity. Cell Physiol Biochem. 54:195–210. 2020.PubMed/NCBI View Article : Google Scholar

113 

Jia S, Agarwal M, Yang J, Horowitz JC, White ES and Kim KK: Discoidin domain receptor 2 signaling regulates fibroblast apoptosis through PDK1/Akt. Am J Respir Cell Mol Biol. 59:295–305. 2018.PubMed/NCBI View Article : Google Scholar

114 

Wang L, Li Z, Wan R, Pan X, Li B, Zhao H, Yang J, Zhao W, Wang S, Wang Q, et al: Single-cell RNA sequencing provides new insights into therapeutic roles of thyroid hormone in idiopathic pulmonary fibrosis. Am J Respir Cell Mol Biol. 69:456–469. 2023.PubMed/NCBI View Article : Google Scholar

115 

Adams TS, Schupp JC, Poli S, Ayaub EA, Neumark N, Ahangari F, Chu SG, Raby BA, DeIuliis G, Januszyk M, et al: Single-cell RNA-seq reveals ectopic and aberrant lung-resident cell populations in idiopathic pulmonary fibrosis. Sci Adv. 6(eaba1983)2020.PubMed/NCBI View Article : Google Scholar

116 

Yang L, Gilbertsen A, Smith K, Xia H, Higgins L, Guerrero C and Henke CA: Proteomic analysis of the IPF mesenchymal progenitor cell nuclear proteome identifies abnormalities in key nodal proteins that underlie their fibrogenic phenotype. Proteomics. 22(e2200018)2022.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

May-2024
Volume 27 Issue 5

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Mei C, Chen T, Huang X, Xiong C, Chen S and Li Y: Role and targeting of the AGC kinase family in pulmonary fibrosis (Review). Exp Ther Med 27: 190, 2024
APA
Mei, C., Chen, T., Huang, X., Xiong, C., Chen, S., & Li, Y. (2024). Role and targeting of the AGC kinase family in pulmonary fibrosis (Review). Experimental and Therapeutic Medicine, 27, 190. https://doi.org/10.3892/etm.2024.12478
MLA
Mei, C., Chen, T., Huang, X., Xiong, C., Chen, S., Li, Y."Role and targeting of the AGC kinase family in pulmonary fibrosis (Review)". Experimental and Therapeutic Medicine 27.5 (2024): 190.
Chicago
Mei, C., Chen, T., Huang, X., Xiong, C., Chen, S., Li, Y."Role and targeting of the AGC kinase family in pulmonary fibrosis (Review)". Experimental and Therapeutic Medicine 27, no. 5 (2024): 190. https://doi.org/10.3892/etm.2024.12478