Next Article in Journal
Pre-Clinical Drug Testing in 2D and 3D Human In Vitro Models of Glioblastoma Incorporating Non-Neoplastic Astrocytes: Tunneling Nano Tubules and Mitochondrial Transfer Modulates Cell Behavior and Therapeutic Response
Next Article in Special Issue
Structural Features of Tight-Junction Proteins
Previous Article in Journal
Usp10 Modulates the Hippo Pathway by Deubiquitinating and Stabilizing the Transcriptional Coactivator Yorkie
Previous Article in Special Issue
Tight Junctions in Cell Proliferation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Reciprocal Association between the Apical Junctional Complex and AMPK: A Promising Therapeutic Target for Epithelial/Endothelial Barrier Function?

1
Laboratory of Biological Science, Graduate School of Frontier Biosciences, Osaka University, Suita, Osaka 565-0811, Japan
2
Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
3
Laboratory of Biological Science, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0811, Japan
4
Strategic Innovation and Research Center, Teikyo University, Tokyo 173-8605, Japan
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2019, 20(23), 6012; https://doi.org/10.3390/ijms20236012
Submission received: 8 November 2019 / Revised: 26 November 2019 / Accepted: 26 November 2019 / Published: 29 November 2019
(This article belongs to the Special Issue The Tight Junction and Its Proteins: More Than Just a Barrier)

Abstract

:
Epithelial/endothelial cells adhere to each other via cell–cell junctions including tight junctions (TJs) and adherens junctions (AJs). TJs and AJs are spatiotemporally and functionally integrated, and are thus often collectively defined as apical junctional complexes (AJCs), regulating a number of spatiotemporal events including paracellular barrier, selective permeability, apicobasal cell polarity, mechano-sensing, intracellular signaling cascades, and epithelial morphogenesis. Over the past 15 years, it has been acknowledged that adenosine monophosphate (AMP)-activated protein kinase (AMPK), a well-known central regulator of energy metabolism, has a reciprocal association with AJCs. Here, we review the current knowledge of this association and show the following evidences: (1) as an upstream regulator, AJs activate the liver kinase B1 (LKB1)–AMPK axis particularly in response to applied junctional tension, and (2) TJ function and apicobasal cell polarization are downstream targets of AMPK and are promoted by AMPK activation. Although molecular mechanisms underlying these phenomena have not yet been completely elucidated, identifications of novel AMPK effectors in AJCs and AMPK-driven epithelial transcription factors have enhanced our knowledge. More intensive studies along this line would eventually lead to the development of AMPK-based therapies, enabling us to manipulate epithelial/endothelial barrier function.

Graphical Abstract

1. Introduction

Epithelial/endothelial cells adhere to each other via cell–cell junctions, thereby collectively functioning as a permselective barrier between the internal and external environments of the body to define the framework of every biological compartment of our body [1,2,3,4,5,6]. Because the size and function of each compartment and their inter-relationships are critical for biological systems, the specific and general principles regulating the permselective barrier function of epithelial/endothelial cells are of considerable interest. Additionally, it is attractive from a therapeutic point of view to investigate methods to manipulate barrier function, given that its disruption can lead to various diseases [7,8,9,10,11,12,13,14,15,16,17]. For example, changes in paracellular barrier function can initiate and accelerate inflammation (both locally and systemically) [10,11,12,15,16,17], contribute to tumor formation [14,18], initiate infection [19], and disrupt ion homeostasis [9,13,20], and have even been suggested as being potentially involved in the initiation and progression of psychiatric and neurodegenerative diseases [21,22,23,24,25,26].
Tight junctions (TJs) are vertebrate-specific junctions that are located most apically in epithelial cell–cell junctions. It is generally accepted that adherens junctions (AJs), which are located immediately at the basal side of TJs, are essential for epithelial cell sheet formation, on the basis of which TJs create a paracellular permselective barrier [6,27,28]. Therefore, TJs and AJs are considered to be spatiotemporally and functionally integrated, and are thus often collectively known as apical junctional complexes (AJCs). In addition to creating a paracellular barrier, AJCs play essential roles in apicobasal cell polarity, mechano-sensing, intracellular signaling cascades, and epithelial morphogenesis [1,6,28,29,30,31,32]. A number of signaling networks are involved in AJC-related events and, over the past 15 years, adenosine monophosphate (AMP)-activated protein kinase (AMPK), a well-known central regulator of energy metabolism signaling pathway, has been shown to have a reciprocal association with AJCs [33,34,35,36,37,38,39,40]. More specifically, AJs activate AMPK particularly in response to applied junctional tension, whereas TJ barrier function and apicobasal cell polarization are promoted by AMPK activation. Herein, we review the critical studies regarding this reciprocal association and discuss the therapeutic potential of AMPK to manipulate epithelial/endothelial barrier function.

2. Overview of Function and Regulation of AMPK

In the 1970s and 1980s, a series of biochemical experiments identified the presence of a 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase kinase, which was found to be activated by AMP [41,42,43]. Meanwhile, an acetyl-coenzyme A (acetyl-CoA) carboxylase kinase was purified from rat liver in the mid-1980s and was also found to be activated by AMP [44]. Later, the independently identified HMG-CoA reductase kinase and acetyl-CoA carboxylase kinase were shown to be the same protein, and this protein was subsequently given the name “AMPK” after its characteristic feature of activation by AMP [45,46].
AMPK is a heterotrimeric serine/threonine kinase composed of one catalytic α-subunit and two regulatory β- and γ-subunits, and is highly conserved in all eukaryotes from protozoa to vertebrates (Figure 1) [47,48,49,50,51,52,53,54,55,56].
AMPK functions as a central regulator of energy metabolism and, once activated, it turns on catabolic processes as well as turns off anabolic processes, leading to upregulation of adenosine triphosphate (ATP) concentrations (Figure 2). Activation of AMPK is determined by the balance of the following three factors (Figure 2) [48,49,57]: (1) the AMP/ATP ratio [48,57], (2) the activity of upstream kinases to phosphorylate α-subunit Thr-172 including liver kinase B1 (LKB1) and Ca2+/calmodulin-dependent protein kinase kinase β (CaMKKβ) [58,59], and (3) the activity of upstream protein phosphatases that dephosphorylate α-subunit Thr-172 such as protein phosphatase 2A and 2C [49,60].
Notably, binding of AMP to the γ-subunit can activate AMPK by three mechanisms that can be antagonized by ATP (Figure 3): (1) AMP drives direct allosteric activation of AMPK [61], (2) AMP renders AMPK susceptible to α-subunit Thr-172 phosphorylation by upstream kinases [62,63], and (3) AMP protects phosphorylated α-subunit Thr-172 from dephosphorylation by upstream phosphatases [64]. It should also be noted that the effect of direct allosteric activation by AMP on AMPK activity is modest (approximately a fivefold increase in activity) [61] compared with that of α-subunit Thr-172 phosphorylation (more than 100-fold increase in activity) [64]; therefore, the phosphorylation status of α-subunit Thr-172 is essentially a molecular on–off switch for AMPK activity [57] (Figure 3). Because AMPK activation has potential to mediate a variety of different cellular consequences, AMPK activation is controlled, not only temporally by the phosphorylation status of α-subunit Thr-172, but also spatially [47,57]. Spatially, AMPK is mobilized to various subcellular organelles only in response to appropriate signals. As examples, glucose starvation leads to AMPK targeting to lysosomes, increased autophagic influx results in AMPK targeting to autophagosomes, and mitochondrial damage causes AMPK targeting to mitochondria [65].

3. Notable Findings in the Mid-2000s Established the Important Role of the LKB1–AMPK Axis in Apicobasal Epithelial Cell Polarity

In 2004, Clevers and colleagues shocked the epithelial cell research community with their seminal paper [33]. Until then, it was believed that apicobasal epithelial cell polarity was established after cells formed cell–cell junctions [66]. However, using intestinal epithelial cell lines with inducible LKB1 activity, they showed that, even in the absence of junctional cell–cell contacts, activation of LKB1 alone can fully establish apicobasal cell polarity in a cell-autonomous manner with the three major aspects of cell polarity all fulfilled, these being the appropriate sorting of apical and basolateral plasma membrane markers, formation of a full brush border, and correct junctional localization of junctional proteins. LKB1 forms a complex with ste-20 related adaptor (STRAD) and mouse protein 25 (MO25) that exerts many functions through the phosphorylation of many substrates [67]. In 2005, the downstream LKB1 effector responsible for establishing apicobasal cell polarity was suggested to be AMPK by investigating the functional consequences of a missense mutation identified in Peutz–Jeghers syndrome (PJS) that is caused by germline mutations of the LKB1 gene [34]. It was found that there is a certain LKB1-C-terminal missense mutation found in PJS patients that abolish the kinase activity responsible for almost all effects of the LKB1/STRAD/MO25 complex but still impair AMPK activity and apicobasal polarity establishment [34]. Subsequently, in 2007, it was found that not only Drosophila LKB1-null mutants but also Drosophila AMPK-null mutants display severe abnormalities in apicobasal cell polarity [36]. These findings collectively showed the crucial role of the LKB1–AMPK axis in apicobasal epithelial cell polarity establishment and have certainly paved the way for subsequent elucidation of the intimate association between AMPK and AJCs.

4. AJs and Junctional Tension Promote the LKB1–AMPK Axis to Provide Energy to Resist Applied Force

Interestingly, in 2009, it was found that AJs can activate LKB1 complexes [38]. In this study, it was shown that active LKB1 complexes co-localize with E-cadherin in polarized Caco-2 and Madin–Darby canine kidney (MDCK) II cells, that E-cadherin-dependent junctional maturation gradually mobilizes LKB1 complexes to AJs, and that AJ localization of LKB1 complexes is critical for the activation of AMPK. However, the physiological relevance of the LKB1–AMPK axis regulation at AJs remained unknown until 2017 when this regulation was shown to be critical for epithelial cells to resist against applied force. In epithelial cells, mechano-sensing, especially at AJs, triggers robust actin cytoskeletal rearrangements to resist externally applied forces [68,69,70]. From an energetic point of view, these changes are very costly, consuming about half of the total ATP in a cell. In 2017, it was found that junctional tension directly applied on E-cadherin, as well as shear stress on MDCK II cells, facilitates AJ-mediated LKB1–AMPK activation. Activated AMPK subsequently increases ATP levels and activates RhoA to support peri-junctional actomyosin filament rearrangement, thereby contributing to the cell stiffness critical for resisting against applied force [39] (Figure 4). In vivo importance of this AJ-mediated LKB1–AMPK regulation is suggested by the study of the tumor suppressor gene folliculin (FLCN), the causative gene for a rare autosomal-dominant disorder named Birt–Hogg–Dubé (BHD) syndrome. It was shown that FLCN deletion in lung epithelium results in downregulation of E-cadherin and LKB1 expression, which leads to the impairment of AMPK activation and alveolar collapse [71]. Collectively, these findings established the critical role of AJs and junctional tension in promoting the LKB1–AMPK axis to resist applied force.

5. TJ Function Is Reinforced by AMPK

The first report that showed the association of AMPK with TJs was published in 2006 [35]. At that time, AMPK was shown to facilitate apicobasal polarity establishment [34,36] and TJs have long been believed to function as a “fence” to prevent the diffusion of proteins and lipids, thereby having a role in apicobasal cell polarity [72] (later, “fence” function of TJs has been basically denied [73,74], although some controversial discussions appeared again recently [75]). Therefore, the role of AMPK in TJ assembly was investigated using a calcium switch experiment, a prevalent experiment to study TJ assembly. In a calcium switch experiment, calcium depletion from the medium of confluent epithelial cells destabilizes AJs, which triggers TJ disassembly with zonula occludens-1 (ZO-1) translocation from cell–cell junctions to the cytoplasm. Calcium restoration causes TJ assembly with ZO-1 redistribution from the cytoplasm to cell–cell junctions to form nascent ZO-1 puncta, which later forms continuous ZO-1 structures. Using this experimental procedure, it was shown that AMPK is activated during TJ assembly in MDCK II cells and that 5-aminoimidazole-4-carboxamide ribonucleoside (AICAR), a potent AMPK activating reagent, facilitates ZO-1 redistribution from the cytoplasm to cell–cell junctions, whereas expression of a dominant-negative AMPK (a kinase-dead form of AMPK) significantly delays ZO-1 redistribution with impaired establishment of the paracellular barrier. It was also noted that AMPK activation alone does not lead to the formation of continuous ZO-1 structures from puncta, suggesting that AMPK is required in the initiation of TJ assembly but other factors are needed to support further TJ maturation [35]. Shortly after this, it was also shown that AMPK activation by AICAR partially protects TJs from disassembly induced by calcium depletion [37]. Altogether, these studies established that TJs are a downstream target of AMPK and are reinforced by its activation.

6. Molecular Basis of the Effects of AMPK on AJCs: Implication of Novel AMPK Effectors in AJCs and AMPK-Driven Epithelial Transcription Factors

Since these early studies, there has been a general consensus that AMPK has a role in reinforcing TJ function and/or establishing apicobasal polarity in a variety of epithelial cells both in vitro and in vivo [76,77,78,79,80,81,82,83,84,85]. Although the molecular mechanisms underlying these phenomena are not yet completely elucidated, recent identification of novel AMPK effectors in AJCs and AMPK-driven epithelial transcription factors have improved our knowledge.
To date, myosin regulatory light chain (MRLC), afadin, cingulin, claudins, and G-alpha interacting vesicle associated protein (GIV)/Girdin were identified as AMPK effectors in AJCs. The first one identified was MRLC, which was originally shown in 2007 to be directly phosphorylated at Thr-21 by AMPK using in vitro phosphorylation assays [36]. In that report, it was shown that this phosphorylation of MRLC is critical in the phenotype of AMPK-null mutants in Drosophila which displays severe abnormalities in apicobasal cell polarity [36]. However, it should be noted that this finding has since been questioned because this phosphorylation site does not match the optimal AMPK substrate motif found in all other established AMPK substrates [86]. In 2011, afadin was identified as the second AMPK substrate in AJCs by the study investigating adhesion-related molecules, which are important in AMPK-dependent TJ assembly [87]. Prior to that study, afadin, together with another cell adhesion molecule nectin, was known to constitute an important adhesion systems in epithelial cells [88], and nectin–afadin-based adhesion was also known to appear during the earliest stages in the process of epithelial morphogenesis prior to the expression of TJ proteins [89]. In that study, it was found that afadin is a direct substrate for AMPK using in vitro phosphorylation assays. It was also found that AMPK activation by AICAR treatment increases the interaction of afadin with ZO-1 and that knocking down afadin expression prevented AMPK-mediated facilitation of TJ assembly in MDCK II cells, suggesting the possibility that AMPK reinforces nectin–afadin-based adhesion at an early stage of cell–cell junction establishment to facilitate TJ assembly [87]. The third AMPK effector in AJCs was identified in 2013, during the search for microtubule-binding proteins in the AJC fraction, when we found that cingulin is a direct AMPK substrate and AMPK-mediated phosphorylation of cingulin facilitates cingulin-mediated linkages of apical microtubule networks to TJs in epithelial cells [80,90]. Cingulin is a specific TJ protein [91] and has two potential evolutionally conserved AMPK target motifs at residues Ser-132 and Ser-150, and we showed that wild-type cingulin can be directly phosphorylated by AMPK at both sites using in vitro phosphorylation assays. Furthermore, we showed that AMPK-mediated cingulin phosphorylation strengthens the interaction of cingulin with microtubules in mouse mammary gland epithelial Eph4 cells and is important in epithelial three-dimensional morphogenesis [80,92]. The fourth group of AMPK effectors in AJCs is the claudins. In 2014, it was shown that AMPK activation by AICAR increases phosphorylation of claudin-4 at Ser-199 in an extracellular signal-regulated kinase 1/2-dependent manner in SV40 immortalized rat submandibular acinar cell lines [93]. Shortly after this, it was shown that claudin-1 can also be directly phosphorylated at Thr-191 by AMPK using in vitro phosphorylation assays and that this phosphorylation may potentially be important in TJ reinforcement in epithelial Eph4 cells [94]. The last AMPK effector in AJCs discovered so far was GIV/Girdin, which was identified as a direct AMPK substrate in 2016 [95]. Prior to that work, GIV/Girdin was shown to be a guanine-nucleotide-exchange factor (GEF) for Gαi3 in trimeric G proteins and to play an important role in TJ formation and establishment of apicobasal epithelial cell polarity [96]. In that work, it was found that GIV/Girdin has an evolutionally conserved, potential AMPK phosphorylation motif at Ser-245 in the N-terminus of GIV/Girdin and that AMPK directly phosphorylates GIV/Girdin at Ser-245 using in vitro phosphorylation assays. It was also shown that, during apicobasal cell polarity establishment in MDCK II cells, GIV/Girdin becomes phosphorylated by AMPK and subsequently localizes at TJs through its binding affinity for the TJ-linked apical microtubule network. Further, it was shown that this phosphorylation of GIV/Girdin is essential in AMPK-dependent TJ formation and apicobasal epithelial cell polarity establishment, and that an oncogenic mutant of GIV which cannot be phosphorylated by AMPK increases tumor cell growth through the defects in establishing apicobasal cell polarity [95]. These results collectively show that there are several AMPK substrates in AJCs (MRLC, cingulin, claudins, and GIV/Girdin) and that the phosphorylation of these substrates impacts TJ function and apicobasal cell polarity, although how AMPK coordinates these different pathways merits future investigation (Figure 5).
Another important mechanism explaining the association between AMPK and TJs was described in 2017 [97]. Prior studies suggested the possibility that AMPK upregulates the expression of TJ proteins, thereby exerting a positive effect on TJ function; however, the underlying mechanism remained unknown [81,84]. In that study, it was found that, in intestinal epithelial Caco-2 cells, AMPK activation by AICAR upregulates expression of caudal type homeobox 2 (CDX2), a key transcription factor governing the differentiation of intestinal epithelial cells. It was also shown that this upregulation is caused by AMPK-dependent recruitment of methylase PRC2 and demethylase LSD1 to induce histone modifications of the CDX2 promoter. Furthermore, it was shown that intestine-conditional AMPK-knockout mice displayed impairment of intestinal epithelial differentiation and barrier function with a significant reduction in CDX2 expression [97]. Subsequent research found that AMPK activation by purple potato extract (PPE) in Caco-2 cells upregulates TJ protein expression in correlation with CDX2 expression and that AMPK-knockout abolishes the positive effects of PPE on CDX2 expression and TJ protein expression [98]. These findings suggest that, in addition to the AMPK substrates in AJCs, an AMPK-driven epithelial transcription factor plays an important role in AMPK-dependent TJ reinforcement via upregulation of TJ protein content (Figure 6).

7. Role of AMPK in Endothelial Barrier Function

Thus far, we have mainly focused on the role of AMPK in epithelial AJCs. Another intriguing aspect of AMPK is that it also has been shown to enhance endothelial TJ function [99,100,101,102]. An endothelial cell monolayer lines the inside of the intima of all blood and lymphatic vessels, forming a semipermeable barrier between the interstitial fluids within tissues and circulating fluids [103]. In general, TJs together with AJs form pericellular zipper-like structures between endothelial cells to restrict free diffusion of molecules, but the specific architecture and composition varies in different vascular beds. For example, TJs are more sophisticated in small arterioles, especially in highly specialized vascular beds like the blood–brain barrier (BBB) and inner blood–retinal barrier, whereas AJs are more prominent in postcapillary venules [103,104,105].
The overall molecular composition of endothelial TJs and AJs are similar to those of epithelial cells, with some small differences [103]. For example, the cadherin-family protein in epithelial AJs is E-cadherin, whereas it is vascular endothelial-cadherin (VE-cadherin) in endothelial AJs. It is noteworthy that N-cadherin is another major cadherin-family protein expressed in endothelial cells, but is excluded from endothelial AJs for an unknown reason [106,107]. N-cadherin has been shown to be important in the interactions of endothelial cells with pericytes and smooth muscle cells, which are thought to play an essential role in the maturation and stabilization of endothelial cells [108,109]. Interestingly, in 2011, using a pulmonary microvascular endothelial cell (PMVEC) wound healing model, it was found that AMPK-mediated TJ reinforcement is critical for PMVEC repair and that AMPK colocalizes with N-cadherin in PMVEC [99]. Following this finding, the biological importance of the AMPK–N-cadherin interaction was investigated using the same PMVEC wound healing model, and it was found that a short-hairpin RNA which decreases the expression of N-cadherin causes translocation of AMPK away from the membrane and inhibits AMPK-mediated barrier repair [110]. These findings collectively suggest that the N-cadherin–AMPK axis may be important for endothelial barrier assembly, but further studies are warranted for validation of these intriguing findings.

8. Therapeutic Potential of Targeting AMPK for Manipulation of Epithelial Barrier Function

One of the fascinating features of AMPK is that it is “druggable”. Indeed, we can artificially activate AMPK either directly (direct activator) or indirectly through AMP or calcium accumulation (indirect activator) using preexisting food compounds such as polyphenols and α-lipoic acid, as well as drugs such as metformin, thiazolidinediones, AICAR, and A79662 (summarized in Table 1). This leads to the question of whether these agents have any potential as therapeutics for diseases caused by barrier dysfunction. The answer may be “yes”.
By improving intestinal epithelial barrier dysfunction, metformin-induced activation of AMPK can alleviate inflammatory bowel disease in mouse models induced by interleukin-10 deficiency, dextran sulfate sodium, and heat stress [84,85,124]. Metformin-induced intestinal barrier reinforcement was also shown to alleviate fructose-induced non-alcoholic fatty liver disease by blocking translocation of bacterial endotoxin [78]. Moreover, it was shown that metformin-induced AMPK activation may inhibit respiratory infection via reinforcement of airway epithelial barriers [79], attenuate the severity of ischemia-induced renal tubular injury by facilitating TJ assembly in renal epithelial cells [77], and facilitate recovery from acute ischemic stroke by attenuating BBB disruption [102,125] (metformin is an indirect AMPK activator that has other AMPK-independent effects; however, these protective effects of metformin on epithelial barrier function was shown to be dependent on AMPK [77,78,79,84,85,102,124,125]). Furthermore, AICAR-induced AMPK activation was also shown to alleviate lipopolysaccharide (LPS)-induced lung injury via restoring pulmonary endothelial barrier integrity [110], and LPS-induced cardiac dysfunction by attenuating left ventricular wall edema [100]. Therefore, these results collectively show that AMPK is a promising therapeutic target for epithelial/endothelial barrier manipulation. However, it should be noted that AMPK has wide variety of function in wide variety of organs with its activity being spatiotemporally regulated. Therefore, systemic and uniform AMPK activation would cause too many off-target effects. AMPK subunits (α, β, or γ) occur as multiple isoforms encoded by distinct genes (α1 or α2; β1 or β2; γ1, γ2, or γ3), giving rise to 12 possible AMPK α/β/γ heterotrimers that are differently expressed in different tissues and exert different effects [47]. For example, the α1/β2/γ1 heterotrimer is predominant in intestinal epithelial cells [126] and, while α1/β2/γ1, α2/β2/γ1, and α1/β2/γ3 heterotrimers are present in skeletal muscles [127,128], only the α2/β2/γ3 heterotrimer is activated during high intensity exercise [128,129]. Most AMPK activators/inhibitors that are commonly used today are not isoform-specific; therefore, further elucidation of isoform specific localization and/or function, as well as further development of isoform-specific activators and inhibitors, is important for AMPK-based organ- and/or barrier-specific therapy.

9. Conclusions

AMPK is a well-known central regulator of energy metabolism. As we have reviewed here, it has also been shown over 15 years that AMPK has a reciprocal association with AJCs, and fine-tunes TJ function and apicobasal cell polarity establishment in epithelial/endothelial cells. The complete molecular mechanisms underlying these phenomena have not yet been elucidated; however, identifications of novel AMPK effectors in AJCs and AMPK-driven epithelial transcription factors have enhanced our knowledge, and recent technological advances will contribute to better understanding. For example, recent development of transgenic mice expressing a highly sensitive fluorescence resonance energy transfer-based biosensor would visualize dynamic AMPK responses in vivo [130,131]. The formation and regulation of AJC-mediated epithelial cell–cell adhesion and paracellular barrier are critical for the biological systems with its disruption being implicated in various diseases; therefore, further elucidation of molecular details underlying an association between AMPK and AJCs will contribute to the better understanding of epithelial cell physiology as well as the development of therapies, enabling us to manipulate epithelial/endothelial barrier function.

Author Contributions

K.T., writing—original draft preparation; T.Y., A.T., and S.T., writing—review and editing.

Funding

This work was supported by Core Research for Evolutionary Science and Technology (CREST) of the Japan Science and Technology Agency (JST) (to Sachiko Tsukita), Grant-in-Aid for Specially Promoted Research (JP19H05468, to Sachiko Tsukita), Grant-in-Aid for Early-Career Scientists (JP18K14696, to Tomoki Yano), and Grant-in-Aid for Scientific Research (B) (JP16H05121, to Atsushi Tamura) from Japan Society for the Promotion of Science (JSPS) of Japan.

Acknowledgments

We thank all members of Tsukita Lab for their useful comments and discussions.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

TJsTight junctions
AJsAdherens junctions
AMPAdenosine monophosphate
AMPKAMP-activated protein kinase
HMG-CoA3-hydroxy-3-methylglutaryl-coenzyme A
acetyl-CoAAcetyl-coenzyme A
ATPAdenosine triphosphate
LKB1Liver kinase B1
CaMKKβCa2+/calmodulin-dependent protein kinase kinase β
PJSPeutz–Jeghers syndrome
MDCKMadin–Darby canine kidney
FLCNFolliculin
BHDBirt–Hogg–Dubé
AICAR5-Aminoimidazole-4-carboxamide ribonucleoside
MRLCMyosin regulatory light chain
GIVG-alpha interacting vesicle associated protein
GEFGuanine-nucleotide-exchange factor
CDX2Caudal type homeobox 2
PPEPurple potato extract
BBBBlood–brain barrier
VE-cadherinvascular endothelial-cadherin
PMVECPulmonary microvascular endothelial cell
ZMP5-amino-1-β-d-ribofuranosylimidazole- 4-carboxamide-5′-monophosphate
LPSLipopolysaccharide

References

  1. Tsukita, S.; Furuse, M.; Itoh, M. Multifunctional strands in tight junctions. Nat. Rev. Mol. Cell Biol. 2001, 2, 285–293. [Google Scholar] [CrossRef] [PubMed]
  2. Tamura, A.; Yamazaki, Y.; Hayashi, D.; Suzuki, K.; Sentani, K.; Yasui, W.; Tsukita, S. Claudin-based paracellular proton barrier in the stomach. Ann. N. Y. Acad. Sci. 2012, 1258, 108–114. [Google Scholar] [CrossRef] [PubMed]
  3. Tamura, A.; Tsukita, S. Paracellular barrier and channel functions of TJ claudins in organizing biological systems: Advances in the field of barriology revealed in knockout mice. Semin. Cell Dev. Biol. 2014, 36, 177–185. [Google Scholar] [CrossRef] [PubMed]
  4. Suzuki, H.; Tani, K.; Tamura, A.; Tsukita, S.; Fujiyoshi, Y. Model for the architecture of claudin-based paracellular ion channels through tight junctions. J. Mol. Biol. 2015, 427, 291–297. [Google Scholar] [CrossRef]
  5. Nakamura, S.; Irie, K.; Tanaka, H.; Nishikawa, K.; Suzuki, H.; Saitoh, Y.; Tamura, A.; Tsukita, S.; Fujiyoshi, Y. Morphologic determinant of tight junctions revealed by claudin-3 structures. Nat. Commun. 2019, 10, 816. [Google Scholar] [CrossRef]
  6. Tsukita, S.; Tanaka, H.; Tamura, A. The Claudins: From Tight Junctions to Biological Systems. Trends Biochem. Sci. 2019, 44, 141–152. [Google Scholar] [CrossRef]
  7. Tanaka, H.; Tamura, A.; Suzuki, K.; Tsukita, S. Site-specific distribution of claudin-based paracellular channels with roles in biological fluid flow and metabolism. Ann. N. Y. Acad. Sci. 2017, 1405, 44–52. [Google Scholar] [CrossRef]
  8. Wada, M.; Tamura, A.; Takahashi, N.; Tsukita, S. Loss of claudins 2 and 15 from mice causes defects in paracellular Na+ flow and nutrient transport in gut and leads to death from malnutrition. Gastroenterology 2013, 144, 369–380. [Google Scholar] [CrossRef]
  9. Matsumoto, K.; Imasato, M.; Yamazaki, Y.; Tanaka, H.; Watanabe, M.; Eguchi, H.; Nagano, H.; Hikita, H.; Tatsumi, T.; Takehara, T.; et al. Claudin 2 deficiency reduces bile flow and increases susceptibility to cholesterol gallstone disease in mice. Gastroenterology 2014, 147, 1134–1145.e10. [Google Scholar] [CrossRef]
  10. Tanaka, H.; Takechi, M.; Kiyonari, H.; Shioi, G.; Tamura, A.; Tsukita, S. Intestinal deletion of Claudin-7 enhances paracellular organic solute flux and initiates colonic inflammation in mice. Gut 2015, 64, 1529–1538. [Google Scholar] [CrossRef]
  11. Tokumasu, R.; Yamaga, K.; Yamazaki, Y.; Murota, H.; Suzuki, K.; Tamura, A.; Bando, K.; Furuta, Y.; Katayama, I.; Tsukita, S. Dose-dependent role of claudin-1 in vivo in orchestrating features of atopic dermatitis. Proc. Natl. Acad. Sci. USA 2016, 113, E4061–E4068. [Google Scholar] [CrossRef] [PubMed]
  12. Citi, S. Intestinal barriers protect against disease. Science 2018, 359, 1097–1098. [Google Scholar] [CrossRef] [PubMed]
  13. Tanaka, H.; Imasato, M.; Yamazaki, Y.; Matsumoto, K.; Kunimoto, K.; Delpierre, J.; Meyer, K.; Zerial, M.; Kitamura, N.; Watanabe, M.; et al. Claudin-3 regulates bile canalicular paracellular barrier and cholesterol gallstone core formation in mice. J. Hepatol. 2018, 69, 1308–1316. [Google Scholar] [CrossRef] [PubMed]
  14. Suzuki, K.; Sentani, K.; Tanaka, H.; Yano, T.; Suzuki, K.; Oshima, M.; Yasui, W.; Tamura, A.; Tsukita, S. Deficiency of Stomach-Type Claudin-18 in Mice Induces Gastric Tumor Formation Independent of H pylori Infection. Cell. Mol. Gastroenterol. Hepatol. 2019, 8, 119–142. [Google Scholar] [CrossRef]
  15. Nalle, S.C.; Zuo, L.; Ong, M.L.D.M.; Singh, G.; Worthylake, A.M.; Choi, W.; Manresa, M.C.; Southworth, A.P.; Edelblum, K.L.; Baker, G.J.; et al. Graft-versus-host disease propagation depends on increased intestinal epithelial tight junction permeability. J. Clin. Investig. 2019, 129, 902–914. [Google Scholar] [CrossRef]
  16. Graham, W.V.; He, W.; Marchiando, A.M.; Zha, J.; Singh, G.; Li, H.-S.; Biswas, A.; Ong, M.L.D.M.; Jiang, Z.-H.; Choi, W.; et al. Intracellular MLCK1 diversion reverses barrier loss to restore mucosal homeostasis. Nat. Med. 2019, 25, 690–700. [Google Scholar] [CrossRef]
  17. Kuo, W.-T.; Shen, L.; Zuo, L.; Shashikanth, N.; Ong, M.L.D.M.; Wu, L.; Zha, J.; Edelblum, K.L.; Wang, Y.; Wang, Y.; et al. Inflammation-induced Occludin Downregulation Limits Epithelial Apoptosis by Suppressing Caspase-3 Expression. Gastroenterology 2019, 157, 1323–1337. [Google Scholar] [CrossRef]
  18. Hagen, S.J.; Ang, L.-H.; Zheng, Y.; Karahan, S.N.; Wu, J.; Wang, Y.E.; Caron, T.J.; Gad, A.P.; Muthupalani, S.; Fox, J.G. Loss of Tight Junction Protein Claudin 18 Promotes Progressive Neoplasia Development in Mouse Stomach. Gastroenterology 2018, 155, 1852–1867. [Google Scholar] [CrossRef]
  19. Shah, J.; Rouaud, F.; Guerrera, D.; Vasileva, E.; Popov, L.M.; Kelley, W.L.; Rubinstein, E.; Carette, J.E.; Amieva, M.R.; Citi, S. A Dock-and-Lock Mechanism Clusters ADAM10 at Cell-Cell Junctions to Promote α-Toxin Cytotoxicity. Cell Rep. 2018, 25, 2132–2147.e7. [Google Scholar] [CrossRef]
  20. King, A.J.; Siegel, M.; He, Y.; Nie, B.; Wang, J.; Koo-McCoy, S.; Minassian, N.A.; Jafri, Q.; Pan, D.; Kohler, J.; et al. Inhibition of sodium/hydrogen exchanger 3 in the gastrointestinal tract by tenapanor reduces paracellular phosphate permeability. Sci. Transl. Med. 2018, 10, eaam6474. [Google Scholar] [CrossRef]
  21. Clairembault, T.; Leclair-Visonneau, L.; Coron, E.; Bourreille, A.; Le Dily, S.; Vavasseur, F.; Heymann, M.-F.; Neunlist, M.; Derkinderen, P. Structural alterations of the intestinal epithelial barrier in Parkinson’s disease. Acta Neuropathol. Commun. 2015, 3, 12. [Google Scholar] [CrossRef]
  22. Montagne, A.; Zhao, Z.; Zlokovic, B.V. Alzheimer’s disease: A matter of blood-brain barrier dysfunction? J. Exp. Med. 2017, 214, 3151–3169. [Google Scholar] [CrossRef] [PubMed]
  23. Greene, C.; Kealy, J.; Humphries, M.M.; Gong, Y.; Hou, J.; Hudson, N.; Cassidy, L.M.; Martiniano, R.; Shashi, V.; Hooper, S.R.; et al. Dose-dependent expression of claudin-5 is a modifying factor in schizophrenia. Mol. Psychiatry 2018, 23, 2156–2166. [Google Scholar] [CrossRef] [PubMed]
  24. Tsukita, K.; Taguchi, T.; Sakamaki-Tsukita, H.; Tanaka, K.; Suenaga, T. The vagus nerve becomes smaller in patients with Parkinson’s disease: A preliminary cross-sectional study using ultrasonography. Parkinsonism Relat. Disord. 2018, 55, 148–149. [Google Scholar] [CrossRef] [PubMed]
  25. Tsukita, K.; Sakamaki-Tsukita, H.; Tanaka, K.; Suenaga, T.; Takahashi, R. Value of in vivo α-synuclein deposits in Parkinson’s disease: A systematic review and meta-analysis. Mov. Disord. 2019, 34, 1452–1463. [Google Scholar] [CrossRef] [PubMed]
  26. Van IJzendoorn, S.C.D.; Derkinderen, P. The Intestinal Barrier in Parkinson’s Disease: Current State of Knowledge. J. Parkinsons Dis. 2019, 9, S323–S329. [Google Scholar] [CrossRef] [PubMed]
  27. Rosenthal, R.; Günzel, D.; Theune, D.; Czichos, C.; Schulzke, J.-D.; Fromm, M. Water channels and barriers formed by claudins. Ann. N. Y. Acad. Sci. 2017, 1397, 100–109. [Google Scholar] [CrossRef] [PubMed]
  28. Konishi, S.; Yano, T.; Tanaka, H.; Mizuno, T.; Kanoh, H.; Tsukita, K.; Namba, T.; Tamura, A.; Yonemura, S.; Gotoh, S.; et al. Vinculin is critical for the robustness of the epithelial cell sheet paracellular barrier for ions. Life Sci. Alliance 2019, 2, e201900414. [Google Scholar] [CrossRef]
  29. Yonemura, S.; Wada, Y.; Watanabe, T.; Nagafuchi, A.; Shibata, M. alpha-Catenin as a tension transducer that induces adherens junction development. Nat. Cell Biol. 2010, 12, 533–542. [Google Scholar] [CrossRef]
  30. Zihni, C.; Mills, C.; Matter, K.; Balda, M.S. Tight junctions: From simple barriers to multifunctional molecular gates. Nat. Rev. Mol. Cell Biol. 2016, 17, 564–580. [Google Scholar] [CrossRef]
  31. Yano, T.; Kanoh, H.; Tamura, A.; Tsukita, S. Apical cytoskeletons and junctional complexes as a combined system in epithelial cell sheets. Ann. N. Y. Acad. Sci. 2017, 1405, 32–43. [Google Scholar] [CrossRef] [PubMed]
  32. Citi, S. The mechanobiology of tight junctions. Biophys. Rev. In press. [CrossRef] [PubMed]
  33. Baas, A.F.; Kuipers, J.; van der Wel, N.N.; Batlle, E.; Koerten, H.K.; Peters, P.J.; Clevers, H.C. Complete polarization of single intestinal epithelial cells upon activation of LKB1 by STRAD. Cell 2004, 116, 457–466. [Google Scholar] [CrossRef] [Green Version]
  34. Forcet, C.; Etienne-Manneville, S.; Gaude, H.; Fournier, L.; Debilly, S.; Salmi, M.; Baas, A.; Olschwang, S.; Clevers, H.; Billaud, M. Functional analysis of Peutz-Jeghers mutations reveals that the LKB1 C-terminal region exerts a crucial role in regulating both the AMPK pathway and the cell polarity. Hum. Mol. Genet. 2005, 14, 1283–1292. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Zhang, L.; Li, J.; Young, L.H.; Caplan, M.J. AMP-activated protein kinase regulates the assembly of epithelial tight junctions. Proc. Natl. Acad. Sci. USA 2006, 103, 17272–17277. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Lee, J.H.; Koh, H.; Kim, M.; Kim, Y.; Lee, S.Y.; Karess, R.E.; Lee, S.-H.; Shong, M.; Kim, J.-M.; Kim, J.; et al. Energy-dependent regulation of cell structure by AMP-activated protein kinase. Nature 2007, 447, 1017–1020. [Google Scholar] [CrossRef]
  37. Zheng, B.; Cantley, L.C. Regulation of epithelial tight junction assembly and disassembly by AMP-activated protein kinase. Proc. Natl. Acad. Sci. USA 2007, 104, 819–822. [Google Scholar] [CrossRef] [Green Version]
  38. Sebbagh, M.; Santoni, M.-J.; Hall, B.; Borg, J.-P.; Schwartz, M.A. Regulation of LKB1/STRAD localization and function by E-cadherin. Curr. Biol. 2009, 19, 37–42. [Google Scholar] [CrossRef] [Green Version]
  39. Bays, J.L.; Campbell, H.K.; Heidema, C.; Sebbagh, M.; DeMali, K.A. Linking E-cadherin mechanotransduction to cell metabolism through force-mediated activation of AMPK. Nat. Cell Biol. 2017, 19, 724–731. [Google Scholar] [CrossRef] [Green Version]
  40. Rowart, P.; Wu, J.; Caplan, M.J.; Jouret, F. Implications of AMPK in the Formation of Epithelial Tight Junctions. Int. J. Mol. Sci. 2018, 19, 2040. [Google Scholar] [CrossRef] [Green Version]
  41. Beg, Z.H.; Allmann, D.W.; Gibson, D.M. Modulation of 3-hydroxy-3-methylglutaryl coenzyme A reductase activity with cAMP and wth protein fractions of rat liver cytosol. Biochem. Biophys. Res. Commun. 1973, 54, 1362–1369. [Google Scholar] [CrossRef]
  42. Carlson, C.A.; Kim, K.H. Regulation of hepatic acetyl coenzyme A carboxylase by phosphorylation and dephosphorylation. J. Biol. Chem. 1973, 248, 378–380. [Google Scholar] [CrossRef]
  43. Ferrer, A.; Caelles, C.; Massot, N.; Hegardt, F.G. Activation of rat liver cytosolic 3-hydroxy-3-methylglutaryl coenzyme A reductase kinase by adenosine 5′-monophosphate. Biochem. Biophys. Res. Commun. 1985, 132, 497–504. [Google Scholar] [CrossRef]
  44. Carling, D.; Zammit, V.A.; Hardie, D.G. A common bicyclic protein kinase cascade inactivates the regulatory enzymes of fatty acid and cholesterol biosynthesis. FEBS Lett. 1987, 223, 217–222. [Google Scholar] [CrossRef] [Green Version]
  45. Sim, A.T.; Hardie, D.G. The low activity of acetyl-CoA carboxylase in basal and glucagon-stimulated hepatocytes is due to phosphorylation by the AMP-activated protein kinase and not cyclic AMP-dependent protein kinase. FEBS Lett. 1988, 233, 294–298. [Google Scholar] [CrossRef] [Green Version]
  46. Grahame Hardie, D.; Carling, D.; Sim, A.T.R. The AMP-activated protein kinase: A multisubstrate regulator of lipid metabolism. Trends Biochem. Sci. 1989, 14, 20–23. [Google Scholar] [CrossRef]
  47. Carling, D. The AMP-activated protein kinase cascade—A unifying system for energy control. Trends Biochem. Sci. 2004, 29, 18–24. [Google Scholar] [CrossRef]
  48. Xiao, B.; Heath, R.; Saiu, P.; Leiper, F.C.; Leone, P.; Jing, C.; Walker, P.A.; Haire, L.; Eccleston, J.F.; Davis, C.T.; et al. Structural basis for AMP binding to mammalian AMP-activated protein kinase. Nature 2007, 449, 496–500. [Google Scholar] [CrossRef]
  49. Xiao, B.; Sanders, M.J.; Underwood, E.; Heath, R.; Mayer, F.V.; Carmena, D.; Jing, C.; Walker, P.A.; Eccleston, J.F.; Haire, L.F.; et al. Structure of mammalian AMPK and its regulation by ADP. Nature 2011, 472, 230–233. [Google Scholar] [CrossRef] [Green Version]
  50. Langendorf, C.G.; Ngoei, K.R.W.; Scott, J.W.; Ling, N.X.Y.; Issa, S.M.A.; Gorman, M.A.; Parker, M.W.; Sakamoto, K.; Oakhill, J.S.; Kemp, B.E. Structural basis of allosteric and synergistic activation of AMPK by furan-2-phosphonic derivative C2 binding. Nat. Commun. 2016, 7, 10912. [Google Scholar] [CrossRef]
  51. Salt, I.P.; Hardie, D.G. AMP-Activated Protein Kinase: An Ubiquitous Signaling Pathway With Key Roles in the Cardiovascular System. Circ. Res. 2017, 120, 1825–1841. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Chen, L.; Jiao, Z.-H.; Zheng, L.-S.; Zhang, Y.-Y.; Xie, S.-T.; Wang, Z.-X.; Wu, J.-W. Structural insight into the autoinhibition mechanism of AMP-activated protein kinase. Nature 2009, 459, 1146–1149. [Google Scholar] [CrossRef] [PubMed]
  53. Li, X.; Wang, L.; Zhou, X.E.; Ke, J.; de Waal, P.W.; Gu, X.; Tan, M.H.E.; Wang, D.; Wu, D.; Xu, H.E.; et al. Structural basis of AMPK regulation by adenine nucleotides and glycogen. Cell Res. 2015, 25, 50–66. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Oakhill, J.S.; Chen, Z.-P.; Scott, J.W.; Steel, R.; Castelli, L.A.; Ling, N.; Macaulay, S.L.; Kemp, B.E. β-Subunit myristoylation is the gatekeeper for initiating metabolic stress sensing by AMP-activated protein kinase (AMPK). Proc. Natl. Acad. Sci. USA 2010, 107, 19237–19241. [Google Scholar] [CrossRef] [Green Version]
  55. Oligschlaeger, Y.; Miglianico, M.; Chanda, D.; Scholz, R.; Thali, R.F.; Tuerk, R.; Stapleton, D.I.; Gooley, P.R.; Neumann, D. The recruitment of AMP-activated protein kinase to glycogen is regulated by autophosphorylation. J. Biol. Chem. 2015, 290, 11715–11728. [Google Scholar] [CrossRef] [Green Version]
  56. Xiao, B.; Sanders, M.J.; Carmena, D.; Bright, N.J.; Haire, L.F.; Underwood, E.; Patel, B.R.; Heath, R.B.; Walker, P.A.; Hallen, S.; et al. Structural basis of AMPK regulation by small molecule activators. Nat. Commun. 2013, 4, 3017. [Google Scholar] [CrossRef] [Green Version]
  57. Carling, D.; Thornton, C.; Woods, A.; Sanders, M.J. AMP-activated protein kinase: New regulation, new roles? Biochem. J. 2012, 445, 11–27. [Google Scholar] [CrossRef] [Green Version]
  58. Hawley, S.A.; Boudeau, J.; Reid, J.L.; Mustard, K.J.; Udd, L.; Mäkelä, T.P.; Alessi, D.R.; Hardie, D.G. Complexes between the LKB1 tumor suppressor, STRAD alpha/beta and MO25 alpha/beta are upstream kinases in the AMP-activated protein kinase cascade. J. Biol. 2003, 2, 28. [Google Scholar] [CrossRef] [Green Version]
  59. Hawley, S.A.; Pan, D.A.; Mustard, K.J.; Ross, L.; Bain, J.; Edelman, A.M.; Frenguelli, B.G.; Hardie, D.G. Calmodulin-dependent protein kinase kinase-beta is an alternative upstream kinase for AMP-activated protein kinase. Cell Metab. 2005, 2, 9–19. [Google Scholar] [CrossRef] [Green Version]
  60. Davies, S.P.; Helps, N.R.; Cohen, P.T.; Hardie, D.G. 5′-AMP inhibits dephosphorylation, as well as promoting phosphorylation, of the AMP-activated protein kinase. Studies using bacterially expressed human protein phosphatase-2C alpha and native bovine protein phosphatase-2AC. FEBS Lett. 1995, 377, 421–425. [Google Scholar]
  61. Cheung, P.C.; Salt, I.P.; Davies, S.P.; Hardie, D.G.; Carling, D. Characterization of AMP-activated protein kinase gamma-subunit isoforms and their role in AMP binding. Biochem. J. 2000, 346, 659–669. [Google Scholar] [CrossRef] [PubMed]
  62. Hong, S.-P.; Leiper, F.C.; Woods, A.; Carling, D.; Carlson, M. Activation of yeast Snf1 and mammalian AMP-activated protein kinase by upstream kinases. Proc. Natl. Acad. Sci. USA 2003, 100, 8839–8843. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Oakhill, J.S.; Steel, R.; Chen, Z.-P.; Scott, J.W.; Ling, N.; Tam, S.; Kemp, B.E. AMPK is a direct adenylate charge-regulated protein kinase. Science 2011, 332, 1433–1435. [Google Scholar] [CrossRef] [PubMed]
  64. Sanders, M.J.; Grondin, P.O.; Hegarty, B.D.; Snowden, M.A.; Carling, D. Investigating the mechanism for AMP activation of the AMP-activated protein kinase cascade. Biochem. J. 2007, 403, 139–148. [Google Scholar] [CrossRef] [PubMed]
  65. Dite, T.A.; Langendorf, C.G.; Hoque, A.; Galic, S.; Rebello, R.J.; Ovens, A.J.; Lindqvist, L.M.; Ngoei, K.R.W.; Ling, N.X.Y.; Furic, L.; et al. AMP-activated protein kinase selectively inhibited by the type II inhibitor SBI-0206965. J. Biol. Chem. 2018, 293, 8874–8885. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Wodarz, A. Establishing cell polarity in development. Nat. Cell Biol. 2002, 4, E39–E44. [Google Scholar] [CrossRef] [PubMed]
  67. Lizcano, J.M.; Göransson, O.; Toth, R.; Deak, M.; Morrice, N.A.; Boudeau, J.; Hawley, S.A.; Udd, L.; Mäkelä, T.P.; Hardie, D.G.; et al. LKB1 is a master kinase that activates 13 kinases of the AMPK subfamily, including MARK/PAR-1. EMBO J. 2004, 23, 833–843. [Google Scholar] [CrossRef] [Green Version]
  68. Daniel, J.L.; Molish, I.R.; Robkin, L.; Holmsen, H. Nucleotide exchange between cytosolic ATP and F-actin-bound ADP may be a major energy-utilizing process in unstimulated platelets. Eur. J. Biochem. 1986, 156, 677–684. [Google Scholar] [CrossRef]
  69. Bernstein, B.W.; Bamburg, J.R. Actin-ATP hydrolysis is a major energy drain for neurons. J. Neurosci. 2003, 23, 1–6. [Google Scholar] [CrossRef]
  70. Borghi, N.; Sorokina, M.; Shcherbakova, O.G.; Weis, W.I.; Pruitt, B.L.; Nelson, W.J.; Dunn, A.R. E-cadherin is under constitutive actomyosin-generated tension that is increased at cell-cell contacts upon externally applied stretch. Proc. Natl. Acad. Sci. USA 2012, 109, 12568–12573. [Google Scholar] [CrossRef] [Green Version]
  71. Goncharova, E.A.; Goncharov, D.A.; James, M.L.; Atochina-Vasserman, E.N.; Stepanova, V.; Hong, S.-B.; Li, H.; Gonzales, L.; Baba, M.; Linehan, W.M.; et al. Folliculin controls lung alveolar enlargement and epithelial cell survival through E-cadherin, LKB1, and AMPK. Cell Rep. 2014, 7, 412–423. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Diamond, J.M. Twenty-first Bowditch lecture. The epithelial junction: Bridge, gate, and fence. Physiologist 1977, 20, 10–18. [Google Scholar] [PubMed]
  73. Umeda, K.; Ikenouchi, J.; Katahira-Tayama, S.; Furuse, K.; Sasaki, H.; Nakayama, M.; Matsui, T.; Tsukita, S.; Furuse, M.; Tsukita, S. ZO-1 and ZO-2 independently determine where claudins are polymerized in tight-junction strand formation. Cell 2006, 126, 741–754. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Ikenouchi, J.; Umeda, K.; Tsukita, S.; Furuse, M.; Tsukita, S. Requirement of ZO-1 for the formation of belt-like adherens junctions during epithelial cell polarization. J. Cell Biol. 2007, 176, 779–786. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Otani, T.; Nguyen, T.P.; Tokuda, S.; Sugihara, K.; Sugawara, T.; Furuse, K.; Miura, T.; Ebnet, K.; Furuse, M. Claudins and JAM-A coordinately regulate tight junction formation and epithelial polarity. J. Cell Biol. 2019, 218, 3372–3396. [Google Scholar] [CrossRef] [Green Version]
  76. Peng, L.; Li, Z.-R.; Green, R.S.; Holzman, I.R.; Lin, J. Butyrate enhances the intestinal barrier by facilitating tight junction assembly via activation of AMP-activated protein kinase in Caco-2 cell monolayers. J. Nutr. 2009, 139, 1619–1625. [Google Scholar] [CrossRef]
  77. Seo-Mayer, P.W.; Thulin, G.; Zhang, L.; Alves, D.S.; Ardito, T.; Kashgarian, M.; Caplan, M.J. Preactivation of AMPK by metformin may ameliorate the epithelial cell damage caused by renal ischemia. Am. J. Physiol. Renal Physiol. 2011, 301, F1346–F1357. [Google Scholar] [CrossRef] [Green Version]
  78. Spruss, A.; Kanuri, G.; Stahl, C.; Bischoff, S.C.; Bergheim, I. Metformin protects against the development of fructose-induced steatosis in mice: Role of the intestinal barrier function. Lab. Investig. 2012, 92, 1020–1032. [Google Scholar] [CrossRef] [Green Version]
  79. Garnett, J.P.; Baker, E.H.; Naik, S.; Lindsay, J.A.; Knight, G.M.; Gill, S.; Tregoning, J.S.; Baines, D.L. Metformin reduces airway glucose permeability and hyperglycaemia-induced Staphylococcus aureus load independently of effects on blood glucose. Thorax 2013, 68, 835–845. [Google Scholar] [CrossRef] [Green Version]
  80. Yano, T.; Matsui, T.; Tamura, A.; Uji, M.; Tsukita, S. The association of microtubules with tight junctions is promoted by cingulin phosphorylation by AMPK. J. Cell Biol. 2013, 203, 605–614. [Google Scholar] [CrossRef] [Green Version]
  81. Park, H.-Y.; Kunitake, Y.; Hirasaki, N.; Tanaka, M.; Matsui, T. Theaflavins enhance intestinal barrier of Caco-2 Cell monolayers through the expression of AMP-activated protein kinase-mediated Occludin, Claudin-1, and ZO-1. Biosci. Biotechnol. Biochem. 2015, 79, 130–137. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Wang, B.; Wu, Z.; Ji, Y.; Sun, K.; Dai, Z.; Wu, G. L-Glutamine Enhances Tight Junction Integrity by Activating CaMK Kinase 2-AMP-Activated Protein Kinase Signaling in Intestinal Porcine Epithelial Cells. J. Nutr. 2016, 146, 501–508. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Patkee, W.R.A.; Carr, G.; Baker, E.H.; Baines, D.L.; Garnett, J.P. Metformin prevents the effects of Pseudomonas aeruginosa on airway epithelial tight junctions and restricts hyperglycaemia-induced bacterial growth. J. Cell. Mol. Med. 2016, 20, 758–764. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Xue, Y.; Zhang, H.; Sun, X.; Zhu, M.-J. Metformin Improves Ileal Epithelial Barrier Function in Interleukin-10 Deficient Mice. PLoS ONE 2016, 11, e0168670. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Chen, L.; Wang, J.; You, Q.; He, S.; Meng, Q.; Gao, J.; Wu, X.; Shen, Y.; Sun, Y.; Wu, X.; et al. Activating AMPK to Restore Tight Junction Assembly in Intestinal Epithelium and to Attenuate Experimental Colitis by Metformin. Front. Pharmacol. 2018, 9, 761. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Shackelford, D.B.; Shaw, R.J. The LKB1-AMPK pathway: Metabolism and growth control in tumour suppression. Nat. Rev. Cancer 2009, 9, 563–575. [Google Scholar] [CrossRef] [PubMed]
  87. Zhang, L.; Jouret, F.; Rinehart, J.; Sfakianos, J.; Mellman, I.; Lifton, R.P.; Young, L.H.; Caplan, M.J. AMP-activated protein kinase (AMPK) activation and glycogen synthase kinase-3β (GSK-3β) inhibition induce Ca2+-independent deposition of tight junction components at the plasma membrane. J. Biol. Chem. 2011, 286, 16879–16890. [Google Scholar] [CrossRef] [Green Version]
  88. Yamada, A.; Fujita, N.; Sato, T.; Okamoto, R.; Ooshio, T.; Hirota, T.; Morimoto, K.; Irie, K.; Takai, Y. Requirement of nectin, but not cadherin, for formation of claudin-based tight junctions in annexin II-knockdown MDCK cells. Oncogene 2006, 25, 5085–5102. [Google Scholar] [CrossRef] [Green Version]
  89. Brakeman, P.R.; Liu, K.D.; Shimizu, K.; Takai, Y.; Mostov, K.E. Nectin proteins are expressed at early stages of nephrogenesis and play a role in renal epithelial cell morphogenesis. Am. J. Physiol. Renal Physiol. 2009, 296, F564–F574. [Google Scholar] [CrossRef]
  90. Yano, T.; Torisawa, T.; Oiwa, K.; Tsukita, S. AMPK-dependent phosphorylation of cingulin reversibly regulates its binding to actin filaments and microtubules. Sci. Rep. 2018, 8, 15550. [Google Scholar] [CrossRef]
  91. Citi, S.; Sabanay, H.; Jakes, R.; Geiger, B.; Kendrick-Jones, J. Cingulin, a new peripheral component of tight junctions. Nature 1988, 333, 272–276. [Google Scholar] [CrossRef] [PubMed]
  92. Mangan, A.J.; Sietsema, D.V.; Li, D.; Moore, J.K.; Citi, S.; Prekeris, R. Cingulin and actin mediate midbody-dependent apical lumen formation during polarization of epithelial cells. Nat. Commun. 2016, 7, 12426. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Xiang, R.-L.; Mei, M.; Cong, X.; Li, J.; Zhang, Y.; Ding, C.; Wu, L.-L.; Yu, G.-Y. Claudin-4 is required for AMPK-modulated paracellular permeability in submandibular gland cells. J. Mol. Cell Biol. 2014, 6, 486–497. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Shiomi, R.; Shigetomi, K.; Inai, T.; Sakai, M.; Ikenouchi, J. CaMKII regulates the strength of the epithelial barrier. Sci. Rep. 2015, 5, 13262. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Aznar, N.; Patel, A.; Rohena, C.C.; Dunkel, Y.; Joosen, L.P.; Taupin, V.; Kufareva, I.; Farquhar, M.G.; Ghosh, P. AMP-activated protein kinase fortifies epithelial tight junctions during energetic stress via its effector GIV/Girdin. Elife 2016, 5, e20795. [Google Scholar] [CrossRef] [Green Version]
  96. Sasaki, K.; Kakuwa, T.; Akimoto, K.; Koga, H.; Ohno, S. Regulation of epithelial cell polarity by PAR-3 depends on Girdin transcription and Girdin-Gαi3 signaling. J. Cell Sci. 2015, 128, 2244–2258. [Google Scholar] [CrossRef] [Green Version]
  97. Sun, X.; Yang, Q.; Rogers, C.J.; Du, M.; Zhu, M.-J. AMPK improves gut epithelial differentiation and barrier function via regulating Cdx2 expression. Cell Death Differ. 2017, 24, 819–831. [Google Scholar] [CrossRef]
  98. Sun, X.; Du, M.; Navarre, D.A.; Zhu, M.-J. Purple Potato Extract Promotes Intestinal Epithelial Differentiation and Barrier Function by Activating AMP-Activated Protein Kinase. Mol. Nutr. Food Res. 2018, 62. [Google Scholar] [CrossRef]
  99. Creighton, J.; Jian, M.; Sayner, S.; Alexeyev, M.; Insel, P.A. Adenosine monophosphate-activated kinase alpha1 promotes endothelial barrier repair. FASEB J. 2011, 25, 3356–3365. [Google Scholar] [CrossRef] [Green Version]
  100. Castanares-Zapatero, D.; Bouleti, C.; Sommereyns, C.; Gerber, B.; Lecut, C.; Mathivet, T.; Horckmans, M.; Communi, D.; Foretz, M.; Vanoverschelde, J.-L.; et al. Connection between cardiac vascular permeability, myocardial edema, and inflammation during sepsis: Role of the α1AMP-activated protein kinase isoform. Crit. Care Med. 2013, 41, e411–e422. [Google Scholar] [CrossRef]
  101. Takata, F.; Dohgu, S.; Matsumoto, J.; Machida, T.; Kaneshima, S.; Matsuo, M.; Sakaguchi, S.; Takeshige, Y.; Yamauchi, A.; Kataoka, Y. Metformin induces up-regulation of blood-brain barrier functions by activating AMP-activated protein kinase in rat brain microvascular endothelial cells. Biochem. Biophys. Res. Commun. 2013, 433, 586–590. [Google Scholar] [CrossRef] [PubMed]
  102. Liu, Y.; Tang, G.; Li, Y.; Wang, Y.; Chen, X.; Gu, X.; Zhang, Z.; Wang, Y.; Yang, G.-Y. Metformin attenuates blood-brain barrier disruption in mice following middle cerebral artery occlusion. J. Neuroinflammation 2014, 11, 177. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Komarova, Y.A.; Kruse, K.; Mehta, D.; Malik, A.B. Protein Interactions at Endothelial Junctions and Signaling Mechanisms Regulating Endothelial Permeability. Circ. Res. 2017, 120, 179–206. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Simionescu, M.; Simionescu, N.; Palade, G.E. Segmental differentiations of cell junctions in the vascular endothelium. The microvasculature. J. Cell Biol. 1975, 67, 863–885. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Schneeberger, E.E. Structure of intercellular junctions in different segments of the intrapulmonary vasculature. Ann. N. Y. Acad. Sci. 1982, 384, 54–63. [Google Scholar] [CrossRef] [PubMed]
  106. Navarro, P.; Ruco, L.; Dejana, E. Differential localization of VE- and N-cadherins in human endothelial cells: VE-cadherin competes with N-cadherin for junctional localization. J. Cell Biol. 1998, 140, 1475–1484. [Google Scholar] [CrossRef] [PubMed]
  107. Gentil-dit-Maurin, A.; Oun, S.; Almagro, S.; Bouillot, S.; Courçon, M.; Linnepe, R.; Vestweber, D.; Huber, P.; Tillet, E. Unraveling the distinct distributions of VE- and N-cadherins in endothelial cells: A key role for p120-catenin. Exp. Cell Res. 2010, 316, 2587–2599. [Google Scholar] [CrossRef]
  108. Kim, J.H.; Kim, J.H.; Yu, Y.S.; Kim, D.H.; Kim, K.-W. Recruitment of pericytes and astrocytes is closely related to the formation of tight junction in developing retinal vessels. J. Neurosci. Res. 2009, 87, 653–659. [Google Scholar] [CrossRef]
  109. Hellström, M.; Gerhardt, H.; Kalén, M.; Li, X.; Eriksson, U.; Wolburg, H.; Betsholtz, C. Lack of pericytes leads to endothelial hyperplasia and abnormal vascular morphogenesis. J. Cell Biol. 2001, 153, 543–553. [Google Scholar] [CrossRef] [Green Version]
  110. Jian, M.-Y.; Liu, Y.; Li, Q.; Wolkowicz, P.; Alexeyev, M.; Zmijewski, J.; Creighton, J. N-cadherin coordinates AMP kinase-mediated lung vascular repair. Am. J. Physiol. Lung Cell Mol. Physiol. 2016, 310, L71–L85. [Google Scholar] [CrossRef] [Green Version]
  111. Zhou, G.; Myers, R.; Li, Y.; Chen, Y.; Shen, X.; Fenyk-Melody, J.; Wu, M.; Ventre, J.; Doebber, T.; Fujii, N.; et al. Role of AMP-activated protein kinase in mechanism of metformin action. J. Clin. Investig. 2001, 108, 1167–1174. [Google Scholar] [CrossRef] [PubMed]
  112. Hawley, S.A.; Ross, F.A.; Chevtzoff, C.; Green, K.A.; Evans, A.; Fogarty, S.; Towler, M.C.; Brown, L.J.; Ogunbayo, O.A.; Evans, A.M.; et al. Use of cells expressing gamma subunit variants to identify diverse mechanisms of AMPK activation. Cell Metab. 2010, 11, 554–565. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Fryer, L.G.D.; Parbu-Patel, A.; Carling, D. The Anti-diabetic drugs rosiglitazone and metformin stimulate AMP-activated protein kinase through distinct signaling pathways. J. Biol. Chem. 2002, 277, 25226–25232. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Ahn, J.; Lee, H.; Kim, S.; Park, J.; Ha, T. The anti-obesity effect of quercetin is mediated by the AMPK and MAPK signaling pathways. Biochem. Biophys. Res. Commun. 2008, 373, 545–549. [Google Scholar] [CrossRef] [PubMed]
  115. Baur, J.A.; Pearson, K.J.; Price, N.L.; Jamieson, H.A.; Lerin, C.; Kalra, A.; Prabhu, V.V.; Allard, J.S.; Lopez-Lluch, G.; Lewis, K.; et al. Resveratrol improves health and survival of mice on a high-calorie diet. Nature 2006, 444, 337–342. [Google Scholar] [CrossRef]
  116. Shen, Q.W.; Zhu, M.J.; Tong, J.; Ren, J.; Du, M. Ca 2+ /calmodulin-dependent protein kinase kinase is involved in AMP-activated protein kinase activation by α-lipoic acid in C2C12 myotubes. Am. J. Physiol. Cell Physiol. 2007, 293, C1395–C1403. [Google Scholar] [CrossRef]
  117. Corton, J.M.; Gillespie, J.G.; Hawley, S.A.; Hardie, D.G. 5-aminoimidazole-4-carboxamide ribonucleoside. A specific method for activating AMP-activated protein kinase in intact cells? Eur. J. Biochem. 1995, 229, 558–565. [Google Scholar] [CrossRef]
  118. Gómez-Galeno, J.E.; Dang, Q.; Nguyen, T.H.; Boyer, S.H.; Grote, M.P.; Sun, Z.; Chen, M.; Craigo, W.A.; van Poelje, P.D.; MacKenna, D.A.; et al. A Potent and Selective AMPK Activator That Inhibits de Novo Lipogenesis. ACS Med. Chem. Lett. 2010, 1, 478–482. [Google Scholar] [CrossRef] [Green Version]
  119. Cool, B.; Zinker, B.; Chiou, W.; Kifle, L.; Cao, N.; Perham, M.; Dickinson, R.; Adler, A.; Gagne, G.; Iyengar, R.; et al. Identification and characterization of a small molecule AMPK activator that treats key components of type 2 diabetes and the metabolic syndrome. Cell Metab. 2006, 3, 403–416. [Google Scholar] [CrossRef] [Green Version]
  120. Calabrese, M.F.; Rajamohan, F.; Harris, M.S.; Caspers, N.L.; Magyar, R.; Withka, J.M.; Wang, H.; Borzilleri, K.A.; Sahasrabudhe, P.V.; Hoth, L.R.; et al. Structural Basis for AMPK Activation: Natural and Synthetic Ligands Regulate Kinase Activity from Opposite Poles by Different Molecular Mechanisms. Structure 2014, 22, 1161–1172. [Google Scholar] [CrossRef] [Green Version]
  121. Zadra, G.; Photopoulos, C.; Tyekucheva, S.; Heidari, P.; Weng, Q.P.; Fedele, G.; Liu, H.; Scaglia, N.; Priolo, C.; Sicinska, E.; et al. A novel direct activator of AMPK inhibits prostate cancer growth by blocking lipogenesis. EMBO Mol. Med. 2014, 6, 519–538. [Google Scholar] [CrossRef] [PubMed]
  122. Handa, N.; Takagi, T.; Saijo, S.; Kishishita, S.; Takaya, D.; Toyama, M.; Terada, T.; Shirouzu, M.; Suzuki, A.; Lee, S.; et al. Structural basis for compound C inhibition of the human AMP-activated protein kinase α2 subunit kinase domain. Acta Crystallogr. D Biol. Crystallogr. 2011, 67, 480–487. [Google Scholar] [CrossRef] [PubMed]
  123. Scott, J.W.; Galic, S.; Graham, K.L.; Foitzik, R.; Ling, N.X.Y.; Dite, T.A.; Issa, S.M.A.; Langendorf, C.G.; Weng, Q.P.; Thomas, H.E.; et al. Inhibition of AMP-Activated Protein Kinase at the Allosteric Drug-Binding Site Promotes Islet Insulin Release. Chem. Biol. 2015, 22, 705–711. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Xia, Z.; Huang, L.; Yin, P.; Liu, F.; Liu, Y.; Zhang, Z.; Lin, J.; Zou, W.; Li, C. L-Arginine alleviates heat stress-induced intestinal epithelial barrier damage by promoting expression of tight junction proteins via the AMPK pathway. Mol. Biol. Rep. in press. [CrossRef] [PubMed] [Green Version]
  125. Farbood, Y.; Sarkaki, A.; Khalaj, L.; Khodagholi, F.; Badavi, M.; Ashabi, G. Targeting Adenosine Monophosphate-Activated Protein Kinase by Metformin Adjusts Post-Ischemic Hyperemia and Extracellular Neuronal Discharge in Transient Global Cerebral Ischemia. Microcirculation 2015, 22, 534–541. [Google Scholar] [CrossRef]
  126. Harmel, E.; Grenier, E.; Bendjoudi Ouadda, A.; El Chebly, M.; Ziv, E.; Beaulieu, J.F.; Sané, A.; Spahis, S.; Laville, M.; Levy, E. AMPK in the small intestine in normal and pathophysiological conditions. Endocrinology 2014, 155, 873–888. [Google Scholar] [CrossRef] [Green Version]
  127. Wojtaszewski, J.F.P.; Birk, J.B.; Frøsig, C.; Holten, M.; Pilegaard, H.; Dela, F. 5′AMP activated protein kinase expression in human skeletal muscle: Effects of strength training and type 2 diabetes. J. Physiol. 2005, 564, 563–573. [Google Scholar] [CrossRef] [Green Version]
  128. Birk, J.B.; Wojtaszewski, J.F.P. Predominant alpha2/beta2/gamma3 AMPK activation during exercise in human skeletal muscle. J. Physiol. 2006, 577, 1021–1032. [Google Scholar] [CrossRef]
  129. Thomson, D.M. The Role of AMPK in the Regulation of Skeletal Muscle Size, Hypertrophy, and Regeneration. Int. J. Mol. Sci. 2018, 19, 3125. [Google Scholar] [CrossRef] [Green Version]
  130. Tsou, P.; Zheng, B.; Hsu, C.-H.; Sasaki, A.T.; Cantley, L.C. A Fluorescent Reporter of AMPK Activity and Cellular Energy Stress. Cell Metab. 2011, 13, 476–486. [Google Scholar] [CrossRef] [Green Version]
  131. Konagaya, Y.; Terai, K.; Hirao, Y.; Takakura, K.; Imajo, M.; Kamioka, Y.; Sasaoka, N.; Kakizuka, A.; Sumiyama, K.; Asano, T.; et al. A Highly Sensitive FRET Biosensor for AMPK Exhibits Heterogeneous AMPK Responses among Cells and Organs. Cell Rep. 2017, 21, 2628–2638. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Domains of adenosine monophosphate (AMP)-activated protein kinase (AMPK) subunits. The kinase domain (KD) in the catalytic α-subunit exerts AMPK kinase activity, and Thr-172 phosphorylation of the KD is critical for its activity. The autoinhibitory domain (AID) binds to the backside of KD, constraining its mobility, thereby inhibiting kinase activity [52]. When AMP is bound at site-3 in the γ-subunit, the α-linker binds to the γ-subunit, thereby pulling AID away from the KD [53]. The C-terminal domain of the α-subunit (α-CTD) is responsible for interacting with the β- and γ-subunits. The allosteric drug and metabolite (ADaM) site at the KD–carbohydrate-binding module (CBM) interface is a binding site for many direct AMPK activators that induces allosteric activation, including A-769662 and compound 991 (see Table 1) [56]. The N-terminal of the regulatory β-subunit has been shown to be myristoylated, which facilitates targeting of AMPK to membranes and is prerequisite for AMP-mediated α-subunit Thr-172 phosphorylation [54]. The CBM in the β-subunit interacts with glycogen, which facilitates targeting of AMPK to glycogen for glycogen turnover regulation [55]. The C-terminal domain of the β-subunit (β-CTD) interacts with both α- and γ- subunits and serves as a connection between the α- and γ- subunits [51]. The regulatory γ-subunits contain four cystathionine beta synthase (CBS) domains, packing together to generate two Bateman domains, thereby providing four potential AMP-binding sites (site-1 to site-4) [48]. However, in mammalian AMPK, only site-1, site-3, and site-4 bind to AMP because aspartic acid residues in a conserved potential binding site in site-2 are changed to arginine [48]. Site-4 contains a permanently-bound AMP molecule, whereas AMP transiently binds to site-1 and site-3 (site-1 has a much stronger binding affinity for AMP than site-3). AMP binding to site-1 causes allosteric activation of AMPK, and AMP binding to site-3 promotes α-subunit Thr-172 phosphorylation by protecting AMPK from upstream phosphatases [49]. Myr, myristoylation.
Figure 1. Domains of adenosine monophosphate (AMP)-activated protein kinase (AMPK) subunits. The kinase domain (KD) in the catalytic α-subunit exerts AMPK kinase activity, and Thr-172 phosphorylation of the KD is critical for its activity. The autoinhibitory domain (AID) binds to the backside of KD, constraining its mobility, thereby inhibiting kinase activity [52]. When AMP is bound at site-3 in the γ-subunit, the α-linker binds to the γ-subunit, thereby pulling AID away from the KD [53]. The C-terminal domain of the α-subunit (α-CTD) is responsible for interacting with the β- and γ-subunits. The allosteric drug and metabolite (ADaM) site at the KD–carbohydrate-binding module (CBM) interface is a binding site for many direct AMPK activators that induces allosteric activation, including A-769662 and compound 991 (see Table 1) [56]. The N-terminal of the regulatory β-subunit has been shown to be myristoylated, which facilitates targeting of AMPK to membranes and is prerequisite for AMP-mediated α-subunit Thr-172 phosphorylation [54]. The CBM in the β-subunit interacts with glycogen, which facilitates targeting of AMPK to glycogen for glycogen turnover regulation [55]. The C-terminal domain of the β-subunit (β-CTD) interacts with both α- and γ- subunits and serves as a connection between the α- and γ- subunits [51]. The regulatory γ-subunits contain four cystathionine beta synthase (CBS) domains, packing together to generate two Bateman domains, thereby providing four potential AMP-binding sites (site-1 to site-4) [48]. However, in mammalian AMPK, only site-1, site-3, and site-4 bind to AMP because aspartic acid residues in a conserved potential binding site in site-2 are changed to arginine [48]. Site-4 contains a permanently-bound AMP molecule, whereas AMP transiently binds to site-1 and site-3 (site-1 has a much stronger binding affinity for AMP than site-3). AMP binding to site-1 causes allosteric activation of AMPK, and AMP binding to site-3 promotes α-subunit Thr-172 phosphorylation by protecting AMPK from upstream phosphatases [49]. Myr, myristoylation.
Ijms 20 06012 g001
Figure 2. Overview of adenosine monophosphate (AMP)-activated protein kinase (AMPK) activation and the biological consequences. Activation of AMPK is determined by the balance of (1) AMP/adenosine triphosphate (ATP) ratio, (2) upstream kinase activity, and (3) upstream phosphatase activity. Once activated, AMPK activates catabolic pathways such as glycolysis, beta-oxidation of fatty acids, and autophagy, as well as suppresses anabolic pathway such as gluconeogenesis and lipid/protein synthesis to increase ATP levels. LKB1, liver kinase B1; STRAD, ste-20 related adaptor; MO25, mouse protein 25; CaMKKβ, CaMKK Ca2+/calmodulin-dependent protein kinase kinase β; PP2A, protein phosphatase 2A; PP2C, protein phosphatase 2C; KD, kinase domain; AID, auto-inhibitory domain; CTD, C-terminal domain; CBM, carbohydrate-binding module; Myr, myristoylation; CBS, cystathionine beta synthase.
Figure 2. Overview of adenosine monophosphate (AMP)-activated protein kinase (AMPK) activation and the biological consequences. Activation of AMPK is determined by the balance of (1) AMP/adenosine triphosphate (ATP) ratio, (2) upstream kinase activity, and (3) upstream phosphatase activity. Once activated, AMPK activates catabolic pathways such as glycolysis, beta-oxidation of fatty acids, and autophagy, as well as suppresses anabolic pathway such as gluconeogenesis and lipid/protein synthesis to increase ATP levels. LKB1, liver kinase B1; STRAD, ste-20 related adaptor; MO25, mouse protein 25; CaMKKβ, CaMKK Ca2+/calmodulin-dependent protein kinase kinase β; PP2A, protein phosphatase 2A; PP2C, protein phosphatase 2C; KD, kinase domain; AID, auto-inhibitory domain; CTD, C-terminal domain; CBM, carbohydrate-binding module; Myr, myristoylation; CBS, cystathionine beta synthase.
Ijms 20 06012 g002
Figure 3. Adenosine monophosphate (AMP) activates AMP-activated protein kinase (AMPK) in several different ways. Allosteric activation results in approximately a fivefold increase in kinase activity, whereas α-subunit Thr-172 phosphorylation causes more than a 100-fold increase in kinase activity; therefore, α-subunit Thr-172 phosphorylation is essentially considered to be a molecular on–off switch for AMPK. KD, kinase domain; AID, auto-inhibitory domain; CTD, C-terminal domain; CBM, carbohydrate-binding module; Myr, myristoylation; CBS, cystathionine beta synthase.
Figure 3. Adenosine monophosphate (AMP) activates AMP-activated protein kinase (AMPK) in several different ways. Allosteric activation results in approximately a fivefold increase in kinase activity, whereas α-subunit Thr-172 phosphorylation causes more than a 100-fold increase in kinase activity; therefore, α-subunit Thr-172 phosphorylation is essentially considered to be a molecular on–off switch for AMPK. KD, kinase domain; AID, auto-inhibitory domain; CTD, C-terminal domain; CBM, carbohydrate-binding module; Myr, myristoylation; CBS, cystathionine beta synthase.
Ijms 20 06012 g003
Figure 4. Adherens junctions (AJs) activate the liver kinase B1 (LKB1)–AMP-activated protein kinase (AMPK) axis. AJs can promote LKB1 complex activation [38], which is facilitated by applied force (zigzag arrows) [39]. Activated LKB1 complexes subsequently activate AMPK, which increases ATP levels and promotes RhoA activation to rearrange cytoskeletons, providing stiffness to resist against applied force.
Figure 4. Adherens junctions (AJs) activate the liver kinase B1 (LKB1)–AMP-activated protein kinase (AMPK) axis. AJs can promote LKB1 complex activation [38], which is facilitated by applied force (zigzag arrows) [39]. Activated LKB1 complexes subsequently activate AMPK, which increases ATP levels and promotes RhoA activation to rearrange cytoskeletons, providing stiffness to resist against applied force.
Ijms 20 06012 g004
Figure 5. AMP-activated protein kinase (AMPK)-mediated regulation of apicobasal polarity establishment and tight junction (TJ) function are at least partially dependent on its phosphorylation of its substrates in AJCs. AMPK has been shown to phosphorylate myosin regulatory light chain (MRLC), cingulin, G-alpha interacting vesicle associated protein (GIV)/Girdin, claudins, and afadin, which can explain the role of AMPK in apicobasal polarity establishment and tight junction function regulation, at least partially. P, phosphate; AJs, adherens junctions.
Figure 5. AMP-activated protein kinase (AMPK)-mediated regulation of apicobasal polarity establishment and tight junction (TJ) function are at least partially dependent on its phosphorylation of its substrates in AJCs. AMPK has been shown to phosphorylate myosin regulatory light chain (MRLC), cingulin, G-alpha interacting vesicle associated protein (GIV)/Girdin, claudins, and afadin, which can explain the role of AMPK in apicobasal polarity establishment and tight junction function regulation, at least partially. P, phosphate; AJs, adherens junctions.
Ijms 20 06012 g005
Figure 6. Overview of the reciprocal association between apical junctional complexes and AMP-activated kinase (AMPK). Adherens junctions (AJs) activate the liver kinase B1 (LKB1)–AMPK axis particularly in response to applied junctional tension. AMPK reinforces tight junction (TJ) function and promotes apicobasal cell polarization establishment through AMPK effectors in AJCs and AMPK-driven epithelial transcription factors.
Figure 6. Overview of the reciprocal association between apical junctional complexes and AMP-activated kinase (AMPK). Adherens junctions (AJs) activate the liver kinase B1 (LKB1)–AMPK axis particularly in response to applied junctional tension. AMPK reinforces tight junction (TJ) function and promotes apicobasal cell polarization establishment through AMPK effectors in AJCs and AMPK-driven epithelial transcription factors.
Ijms 20 06012 g006
Table 1. List of commonly-used AMPK activators and inhibitors.
Table 1. List of commonly-used AMPK activators and inhibitors.
Compound NameMechanismIsoform SelectivityReference
Indirect activatorsMetformin
(biguanides)
Inhibiting the mitochondrial respiratory chain Complex I
→ Increase in AMP/ATP ratio
No[111,112]
Troglitazone (thiazolidinediones)Inhibiting the mitochondrial respiratory chain Complex I
→ Increase in AMP/ATP ratio
No[112,113]
Resveratrol (polyphenols)Inhibiting F1F0–ATPase/ATP synthase
→ Increase in AMP/ATP ratio
No[112,114]
Quercetin (polyphenols)Inhibiting F1F0–ATPase/ATP synthase
→ Increase in AMP/ATP ratio
No[112,115]
α-Lipoic acidIncreasing calcium level?No[116]
Direct activators5-Aminoimidazole-4-carboxamide ribonucleoside (AICAR)Prodrug, converted to 5-amino-1-β-d-ribofuranosylimidazole- 4-carboxamide-5′-monophosphate (ZMP)
→ ZMP binds to site-3 in the γ-subunit
No[112,117]
Compound-13
(C-13)
Prodrug, converted to Compound-2 (C-2)
→ C-2 binds to the γ-subunit
α1 > α2[118]
A-769662Binding to the allosteric drug and metabolite (ADaM) site
→ Allosteric activation
β1 > β2[56,119]
Compound 991Binding to the ADaM site
→ Allosteric activation
β1 > β2[56]
SalicylateBinding to the ADaM site
→ Allosteric activation
β1[120]
MT63-78Binding to the ADaM site
→ Allosteric activation
β1[121]
InhibitorsCompound C (dorsomorphin)Binding to the α-subunit
→ Competitive inhibition of ATP
No[111,122]
MT47-100Binding to the β2-subunit
→ Allosteric inhibition
β2 (β1 activation)[123]
SBI-0206965Binding to the α-subunit
→ Mixed-type inhibition of ATP
No[65]

Share and Cite

MDPI and ACS Style

Tsukita, K.; Yano, T.; Tamura, A.; Tsukita, S. Reciprocal Association between the Apical Junctional Complex and AMPK: A Promising Therapeutic Target for Epithelial/Endothelial Barrier Function? Int. J. Mol. Sci. 2019, 20, 6012. https://doi.org/10.3390/ijms20236012

AMA Style

Tsukita K, Yano T, Tamura A, Tsukita S. Reciprocal Association between the Apical Junctional Complex and AMPK: A Promising Therapeutic Target for Epithelial/Endothelial Barrier Function? International Journal of Molecular Sciences. 2019; 20(23):6012. https://doi.org/10.3390/ijms20236012

Chicago/Turabian Style

Tsukita, Kazuto, Tomoki Yano, Atsushi Tamura, and Sachiko Tsukita. 2019. "Reciprocal Association between the Apical Junctional Complex and AMPK: A Promising Therapeutic Target for Epithelial/Endothelial Barrier Function?" International Journal of Molecular Sciences 20, no. 23: 6012. https://doi.org/10.3390/ijms20236012

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

Article Metrics

Back to TopTop