• <tr id="yyy80"></tr>
  • <sup id="yyy80"></sup>
  • <tfoot id="yyy80"><noscript id="yyy80"></noscript></tfoot>
  • 99热精品在线国产_美女午夜性视频免费_国产精品国产高清国产av_av欧美777_自拍偷自拍亚洲精品老妇_亚洲熟女精品中文字幕_www日本黄色视频网_国产精品野战在线观看 ?

    The Hippo pathway and its correlation with acute kidney injury

    2022-10-17 03:27:32ChiZhangChuanLeiLiKeXinXuZhiHuangZhengGuoZheChengHuiJuanWuJunLiu
    Zoological Research 2022年5期

    Chi Zhang, Chuan-Lei Li, Ke-Xin Xu, Zhi-Huang Zheng, Guo-Zhe Cheng, Hui-Juan Wu, Jun Liu,*

    1 Department of Nephrology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 201600, China

    2 Department of Pathology, School of Basic Medical Science, Fudan University, Shanghai 200030, China

    ABSTRACT Acute kidney injury (AKI) is a significant clinical complication with a substantial impact on morbidity and mortality, for which therapeutic options remain limited. The Hippo signaling pathway is an evolutionarily conserved pathway implicated in cell proliferation, dedifferentiation, and apoptosis via phosphorylation and inactivation of its downstream effectors Yes-associated protein (YAP)/transcriptional co-activator with PDZ-binding motif(TAZ). Recent studies have revealed that the Hippo pathway plays a pivotal role in the pathogenesis and repair of AKI. The Hippo pathway can mediate renal dysfunction through modulation of mitochondrial apoptosis under AKI conditions. Transient activation of YAP/TAZ in the acute phase of AKI may benefit renal recovery and regeneration, whereas persistent activation of YAP/TAZ in severe AKI may lead to maladaptive repair and transition to chronic kidney disease. This review aims to summarize recent findings on the associations between the Hippo pathway and AKI and to identify new therapeutic targets and strategies for AKI.

    Keywords: Hippo pathway; Acute kidney injury(AKI); Maladaptive repair; Fibrosis; Chronic kidney disease (CKD)

    lNTRODUCTlON

    Acute kidney injury (AKI) remains a major medical challenge with high morbidity, mortality, and healthcare costs (Zuk &Bonventre, 2019). Apart from supportive treatment, few preventive and therapeutic options currently exist (Lameire et al., 2013). Among survivors, long-term outcomes of AKI include development of chronic kidney disease (CKD), endstage renal disease (ESRD), and death (Lameire et al., 2013;Venkatachalam et al., 2015; Zuk & Bonventre, 2019).Incomplete recovery from AKI, also called maladaptive repair,can lead to long-term functional deficits and CKD. The progression of adaptive to maladaptive kidney repair in AKI leading to CKD can be attributed to G2/M cell cycle arrest,profibrotic cytokine secretion, myofibroblast generation,extracellular matrix (ECM) production, and inflammation(Ferenbach & Bonventre, 2015; Yu & Bonventre, 2020).

    The mammalian Hippo pathway is a serine/threonine kinase cascade composed of mammalian sterile 20-like 1/2(MST1/2), large tumor suppressor homolog 1/2 (LATS1/2),and Yes-associated protein (YAP)/transcriptional co-activator with PDZ-binding motif (TAZ) (Zhao et al., 2011b). It regulates cell growth and fate determination, organ size, and regeneration through the phosphorylation and inactivation of its downstream effectors YAP/TAZ (Ma et al., 2019). Recent studies have implicated the Hippo pathway in the pathogenesis and repair of AKI and transition to CKD. In this review, we summarize current findings on the correlations between the Hippo pathway and kidney injury and shed some light on future treatment for acute or chronic kidney diseases.

    HlPPO PATHWAY lN MAMMALS: A KlNASE CASCADE

    The Hippo pathway is highly evolutionarily conserved in mammals and consists of a three-step kinase cascade of MST1/2 (also called STK4/3), LATS1/2, and YAP/TAZ (also called WWTR1). Upon Hippo pathway activation, Salvador homolog 1 (Sav1) interacts with and is phosphorylated by MST1/2, thereby acting as a partner of MST1/2 to promote phosphorylation of LATS1/2 (Callus et al., 2006; Tapon et al.,2002). LATS1/2 are activated by MST1/2 through multiple mechanisms: MST1/2 can directly phosphorylate LATS1/2 at the C-terminal hydrophobic motif (Thr1079 for LATS1 and Thr1041 for LATS2), promoting LATS1/2 autophosphorylation at its activation loop. Furthermore, MST1/2-phosphorylated Mps one binder 1 (MOB1) binds to the autoinhibitory domain of LATS1/2, leading to full activation of LATS1/2 (Callus et al.,2006; Chan et al., 2005; Praskova et al., 2004; Wu et al.,2003). Neurofibromin 2 (NF2) can also directly interact with and recruit LATS1/2 to the plasma membrane for phosphorylation by MST1/2 (Yin et al., 2013; Zhang et al.,2010). Activated LATS1/2 then phosphorylate serine on the YAP/TAZ HXRXXS motifs (Hao et al., 2008; Lei et al., 2008;Liu et al., 2010; Zhao et al., 2007, 2010). Phosphorylation of YAP Ser127 and TAZ Ser89 generates 14-3-3 binding sites,and binding to 14-3-3 results in cytoplasmic sequestration(Dong et al., 2007; Zhao et al., 2007). In addition,phosphorylation of YAP Ser381 and TAZ Ser311 leads to subsequent phosphorylation by casein kinase 1 (CK1) and activation of phosphodegron, resulting in the recruitment of SCFbeta-TRCP E3 ubiquitination ligase and proteasomal degradation of both YAP and TAZ (Liu et al., 2010; Zhao et al., 2010).

    When upstream kinases of the Hippo pathway are inactivated, YAP/TAZ are dephosphorylated and activated. As transcriptional co-activators, YAP/TAZ enter the nucleus and bind with TEAD family transcription factors (TEAD 1-4) to induce the expression of target genes (Zhao et al., 2008),such asCTGF,Cyr61,ANKRD1,DIAPH3,amphiregulin, andsurvivin, thereby improving cell proliferation and inhibiting cell death (Bai et al., 2012; Rinschen et al., 2017). Vestigial-like family member 4 (VGLL4) competes with YAP/TAZ for TEAD binding in the nucleus and represses target gene expression(Figure 1) (Jiao et al., 2014; Zhang et al., 2014). The association between the Hippo pathway and AKI has attracted increasing attention, and the effects of Hippo pathway component activation on AKI are summarized in Table 1.

    HlPPO PATHWAY REGULATlON

    Figure 1 Hippo pathway in mammalian cells

    Table 1 Possible role of Hippo pathway component activation in AKl

    In mammalian cells, the Hippo pathway is regulated by a variety of intrinsic and extrinsic factors. Cell polarity, cell-cell junctions, soluble factors, mechanical cues, and metabolic states, such as cellular energy and oxygen stress, can modulate Hippo pathway activity (Boggiano & Fehon, 2012;Meng, 2016; Mo et al., 2015; Zhao et al., 2011b). In particular,mechanical cues, such as the ECM and cellular geometry, are potent regulators (Dupont et al., 2011). For example,YAP/TAZ can be activated when cells are cultured on a matrix with high stiffness or large surface area. In addition, high density of round and compact cells can lead to YAP/TAZ activation. Conversely, low density of flat and loose cells can result in Hippo pathway inactivation, allowing YAP/TAZ to enter the nucleus and promote transcription of target genes(Aragona et al., 2013; Wada et al., 2011). Both apical and basolateral spectrin networks play regulatory roles in the Hippo pathway and function as sensors of mechanical cues(Deng et al., 2015; Fletcher et al., 2015).

    Soluble factors act primarily through G protein-coupled receptors (GPCRs) in the regulation of the Hippo pathway.GPCRs are the largest family of plasma membrane receptors,linking the Hippo pathway to a broad range of upstream signals. GPCR ligands that signal through Gα12/13 or Gαq/11,such as lysophosphatidic acid (LPA), angiotensin II, and thrombin, increase YAP/TAZ nuclear entry by activating Rho GTPases; whereas ligands that signal through Gαs, such as epinephrine and glucagon, can activate LATS1/2 and repress YAP/TAZ via the AC-cAMP-PKA pathway (Yu et al., 2012,2013; Zhou et al., 2015). Rho GTPases and actin cytoskeleton remodeling are key mediators of GPCR ligands and mechanical cues in the regulation of the Hippo pathway(Figure 2) (Dupont et al., 2011; Zhao et al., 2012).

    Figure 2 YAP/TAZ induced negative feedback loop in regulating Hippo pathway homeostasis

    In addition to GPCR ligands, the Hippo pathway also participates in crosstalk with Wingless/Ints (Wnt) (Bernascone& Martin-Belmonte, 2013), transforming growth factor (TGF)-β(Feng et al., 2018b; Seo et al., 2016), bone morphogenetic proteins (BMPs), Notch, Hedgehog (Hh) (Hansen et al., 2015),Src (Kim et al., 2019), Rac-GTPase activating protein 1(RacGAP1) (Zhou et al., 2020), Gsk3β-p53 (Li et al., 2019),and EGF receptor (EGFR)-phosphatidylinositol 3-kinase(Pl3K)-Akt (Chen et al., 2018), which modulate YAP/TAZ activities and collectively control tissue growth and development. The EGFR-PI3K-Akt pathway and RacGAP1 can help renal tubular epithelial cells (RTECs) recover from acute injury by activating YAP (Chen et al., 2018; Zhou et al.,2020), whereas Wnt5a can exacerbate TGF-β1-induced macrophage M2 polarization and renal fibrosis through YAP/TAZ up-regulation (Feng et al., 2018b). These Hippo pathway regulatory factors may provide potential drug targets for the treatment of kidney injury.

    NEGATlVE FEEDBACK LOOP OF HlPPO PATHWAY

    Long-term hyperactivation of YAP/TAZ can lead to massive tissue overgrowth, enhanced stem cell-like properties that promote tumorigenic potential, and increased metastasis by epithelial-mesenchymal transition (EMT) (Harvey et al., 2013;Lei et al., 2008). Therefore, YAP/TAZ activity must be tightly controlled to maintain tissue homeostasis. In mammalian cells,the Hippo pathway exhibits a negative feedback mechanism.YAP/TAZ and TEAD complexes directly induce the transcription of NF2 and LATS2, and indirectly stimulate LATS1/2 kinase activity by increasing NF2 protein abundance.A reciprocal negative regulation also exists between YAP and TAZ dependent on LATS and proteasomal degradation(Moroishi et al., 2015; Park et al., 2016). YAP/TAZ-induced LATS activation can lead to increased angiomotin (AMOT)phosphorylation and protein accumulation. AMOT can directly inhibit YAP/TAZ and indirectly activate LATS, thus contributing to the negative feedback loop (Zhao et al., 2011a).Furthermore, MST1 and AMOT-like 2 (AMOTL2) are upregulated by YAP/TAZ, and AMOTL2 can also activate LATS(Kim et al., 2016; Paramasivam et al., 2011; Park et al., 2016).This negative feedback loop is essential for maintaining proper transient activation of YAP/TAZ in response to stimuli and preventing tumorigenesis. Aberrant increase of OGlcNAcylation on LATS2 can disrupt the LATS-mediated negative feedback loop, leading to YAP/TAZ hyperactivation and carcinogenesis (Kim et al., 2020).

    RENAL DYSFUNCTlON CAN BE EXACERBATED BY HlPPO PATHWAY-MEDlATED MlTOCHONDRlAL FlSSlON FOLLOWlNG AKl

    The kidney is second only to the heart in mitochondrial count and oxygen consumption (Duann & Lin, 2017). Mitochondria undergo significant changes after AKI, which affects the pathophysiology and recovery of renal function (Baligand et al., 2017; Funk & Schnellmann, 2012; Lan et al., 2016).Persistent disruption of mitochondrial homeostasis and sustained tubular damage can result in progression to CKD(Jiang et al., 2020). The pathogenic mechanisms of mitochondrial dysfunction include mitochondrial DNA damage(Lee & Back, 2017), mitophagy disturbance (Duann et al.,2016), and biosynthesis disorder (Xiao et al., 2017). RTECs are rich in mitochondria and the proximal tubule is especially vulnerable to mitochondrial damage (Hall et al., 2009).Mitochondrial apoptosis is considered the main reason for reperfusion-mediated RTEC apoptosis under renal ischemia/reperfusion (I/R) injury, attributable to mitochondrial fission (Perry et al., 2018; Wang et al., 2020b).

    The core proteins that mediate mitochondrial fission and fusion include dynamin-related protein 1 (Drp1), mitofusin(Mfn), and optic atrophy 1 (OPA1). Drp1 mediates mitochondrial fission, whereas Mfn and OPA1 mediate fusion of the outer and inner membranes, respectively (Lee & Yoon,2016). Li et al. (2019) demonstrated that MST1 is markedly up-regulated after renal I/R injury and that higher MST1 expression is associated with Gsk3β-p53 axis activation,which induces mitochondrial fission by Drp1 phosphorylation and F-actin assembly. Mitochondrial fission contributes to mitochondrial potential reduction, reactive oxygen species(ROS) production, mitochondrial permeability transition pore(mPTP) opening, cytochromec(cytc) release, and caspase-9-related mitochondrial apoptosis initiation, leading to RTEC apoptosis and renal dysfunction. Feng et al. (2018a) also showed that MST1 expression is up-regulated in response to renal I/R injury and that genetic ablation of MST1 improves renal function, alleviates reperfusion-mediated RTEC apoptosis, and attenuates kidney vulnerability to I/R injury.Moreover, increased MST1 blocks the AMPK-YAP pathway,thereby diminishing OPA1 expression, repressing mitophagy,and activating mitochondrial apoptosis in reperfused kidneys.Reactivation of the AMPK-YAP-OPA1 signaling pathway provides a survival advantage for RTECs in the context of renal I/R injury by repressing mitochondrial fission (Figure 3).Based on these findings, we speculate that the Hippo pathway plays a vital role in the pathogenesis of AKI and that increased MST1 expression can mediate mitochondrial damage and RTEC apoptosis by modulating mitochondrial fission.

    YAP OVEREXPRESSlON lN RTECs CAN RESULT lN ACQUlRED CYTORESlSTANCE AFTER AKl

    Figure 3 lncrease in YAP and MST1 following AKl can result in RTEC cytoresistance and apoptosis, respectively

    Animals recovering from previous AKI are resistant to subsequent renal challenge with the same agent, termed acquired resistance (Honda et al., 1987). The emergence of acquired cytoresistance after AKI can serve to protect the kidneys from further ischemia or nephrotoxicity (Zager, 2013).This is closely associated with heat shock proteins (Furuya et al., 1997; Mizuno et al., 1997), hypoxia-inducible factor (HIF)-1α (Ma et al., 2009), heme oxygenase-1 (Nath, 2014), cyclindependent kinase inhibitor (CDKI) (Nishioka et al., 2014; Price et al., 2004), growth/proliferation factors (Sun et al., 2011),and DNA repair-related factors (Miyaji et al., 2001; Sano et al.,2000).

    Iwakura et al. (2017) demonstrated that rat renal proximal tubule epithelial cells (RPTCs) that have recovered from uranyl acetate (UA)-induced AKI can develop cytoresistance to subsequent UA treatment. These cells transiently overexpress YAP and its downstream mediatorsurvivin, which may inhibit apoptosis and increase acquired cytoresistance.These effects may be correlated with enhanced G1 arrest via modulation of cell-cycle regulating factors, including cyclin D1,p21, and p27 (Iwakura et al., 2016), and persist until YAP/survivinexpression levels return to baseline (Figure 3).Furthermore, YAP overexpression leads to hyperplasia in RPTCs, while YAP regression leads to a gradual decline in RPTCs with enhanced cell cycle arrest. Based on these findings, considerable attention should be given to the relationship between RTEC cytoresistance and cellular recovery with YAP/survivinexpression in AKI treatment.

    TRANSlENT YAP/TAZ ACTlVATlON PROMOTES RTEC ADAPTlVE REPAlR lN MlLD/MODERATE AKl

    Due to the high abundance of mitochondria, the metabolically active proximal tubule, especially the S3 segment, is a primary target of injury in AKI (Munshi et al., 2011). Renal stem cells can be a cell source for maintaining normal kidney cell turnover (Alexandre et al., 2009; Bruno et al., 2009; Morigi et al., 2008). Mouse kidney progenitor cells (MKPCs) are a unique renal stem cell population, which can contribute to renal repair and survival after ischemic injury (Lee et al.,2010). Notably, intrarenal injection of MKPCs isolated from the medullary and papillary interstitium in ischemic-injured mice can rescue renal damage, with some MKPCs forming vessels with red blood cells inside and some incorporating into renal tubules 7 days after injury. Intrinsic RTEC dedifferentiation,proliferation, and migration can also contribute to renal regeneration after AKI (Duffield & Humphreys, 2011; Lin,2006). Surviving RTECs can regenerate and regain normal kidney function if the injury is mild and transient, but may exhibit maladaptive repair if the injury is severe and recurring,leading to renal fibrosis (Figure 4) (Ferenbach & Bonventre,2015; Qi & Yang, 2018; Yang et al., 2011). Recent studies have demonstrated the pivotal role of the Hippo signaling pathway in RTEC recovery and regeneration after AKI.

    Figure 4 Schematic of Hippo pathway mediating renal repair after AKl

    Chen et al. (2018) reported enhanced YAP expression and nuclear distribution in the RPTCs of kidneys in patients with AKI and mice with I/R AKI. Furthermore, YAP antagonist verteporfin (VP)-treated and RPTC-specific YAP knockout mice both showed more severe kidney injury compared with the controls, indicating that EGFR-Pl3k-Akt signaling mediates YAP activation and subsequent target genecyclin Dandamphiregulin(AREG) expression.Cyclin Dcan induce the phosphorylation of the retinoblastoma protein (Rb), thus activating cyclin-dependent kinase (CDK) 4 and 6, which are important for G1 to S transition during cell proliferation(Hamilton & Infante, 2016; Musgrove et al., 2011). In addition,AREGacts as an EGFR ligand to further initiate EGFR-Aktdependent YAP activation, thereby promoting adaptive repair of the kidney after AKI. Of note, Chen et al. (2018) also suggested that YAP, but not TAZ, is critical for renal recovery from I/R injury as histological recovery is delayed in the YAP/TAZ double knockout mice but not in the TAZ single knockout mice. In contrast, Wu et al. (2020) reported a beneficial role of tubular TAZ in ischemic AKI, inconsistent with the findings of Chen et al. (2018). They demonstrated thatin vivotreatment with chloroquine, which can enhance TAZ expression in RPTCs, preserves renal function, ameliorates tubular injury, and reduces apoptosis in the renal cortex after I/R injury. In contrast, both TAZ antagonist VP treatment and TAZ knockdown exaggerate tubular apoptosis and renal failure after I/R injury. Further studies are essential to clarify the role of TAZ in RTECs after AKI and illustrate the potential therapeutic targets against AKI.

    Transiently enhanced and evenly distributed YAP levels in the cytoplasm and nucleus are reported in mild I/R AKI rat models (Xu et al., 2016). Dedifferentiation markers, including vimentin, α-smooth muscle actin (α-SMA), and proliferatingcell nuclear antigen (PCNA), show similar expression patterns to YAP during recovery after AKI, whereas quiescent tubular markers, including Na+/K+-ATPase and aquaporin 1 (AQP1),are decreased or absent, indicating that YAP may promote the proliferation and redifferentiation of reconstituted epithelia after acute I/R AKI. Moreover, suppression of YAP1 by the N6-methyladenosine mRNA methylase METTL4 promotes renal I/R injury progression (Xu et al., 2020b). Inhibition of YAP/TAZ degradation and nuclear factor-κB (NF-κB)translocation through Rictor/mTORC2 signaling is crucial for preventing renal inflammation and lipopolysaccharide (LPS)-induced AKI, thus revealing the potent effects of YAP/TAZ on alleviation of AKI severity (Figure 4) (Gui et al., 2020).

    PERSlSTENT YAP/TAZ ACTlVATlON lN SEVERE AKl LEADS TO MALADAPTlVE REPAlR AND CKD

    The Hippo pathway may exhibit bidirectional functions during I/R AKI repair. Xu et al. (2016) demonstrated that YAP mainly elicits beneficial effects on the proliferation of injured RTECs during repair, but persistent activation of YAP can impede the redifferentiation of dedifferentiated tubular cells and promote kidney fibrosis. Notably, they found more pronounced YAP activation in rats four weeks after I/R injury in the incomplete repair (I/R 45 min) group than in the complete repair (I/R 30 min) group. Furthermore, connective tissue growth factor(CTGF), fibrotic protein mRNA (including type I, III, and IV collagen), and RTPCs in the G2/M-phase were up-regulated with YAP increase, indicating kidney fibrosis exacerbation. In addition, the incomplete repair (I/R 45 min) group treated with YAP agonist digitoxin showed more severe tubulointerstitial fibrosis (TIF) compared with the control group. Xu et al. (2021)also implicated YAP activation in the post-acute phase of AKI in renal dysfunction and TIF, although it elicits a beneficial effect in the acute phase. Transcription factor KLF4 can cause persistent activation of YAP, while inhibition of the KLF4-YAP pathway can attenuate IR-induced renal function deterioration and slow TIF progress by decreasing the expression levels of TGF-β and CTGF. Zheng et al. (2021) confirmed that severe ischemic AKI can induce tubular maladaptive repair and TIF by persistently activating YAP. YAP activation can further cause monocyte chemoattractant protein 1 (MCP-1)overexpression and promote renal macrophage inflammation,a driving force for progressive TIF and AKI-CKD transition.

    Moreover, down-regulation of YAP can significantly suppress chronic renal fibrosis. Bothin vivoandin vitrostudies report that miR-101a can potentially slow chronic renal fibrosis by blockade of the YAP-TGF-β-Smad signaling pathway via KDM3A (Ding et al., 2020). Kindlin-2 depletion reduces MOB1 degradation and alleviates renal fibrosis via activation of Hippo/YAP signaling in Kindlin-2 knockout mice with unilateral ureteral obstruction (UUO) (Song et al., 2019).Most recently, knockdown of YTHDF1, a modulator of m6A methylation, was shown to alleviate progression of renal fibrosis in both cultured cells induced by TGF-β administration and UUO model mice via down-regulation of YAP (Xing et al.,2022). In conclusion, appropriate modulation of the Hippo signaling pathway, especially intervention in YAP overexpression during repair, may be a potent preventive target in AKI-CKD transition after I/R injury.

    Kidney fibrosis can be induced through Hippo pathwaymediated EMT

    The severity and frequency of RPTC injury can determine whether the repair mechanism leads to complete recovery or maladaptive repair (Takaori et al., 2016). Maladaptive repair,i.e., renal fibrosis, is characterized by cell cycle arrest at the G2/M phase and a senescence-associated secretory phenotype (SASP). RTECs may turn to a mesenchymal phenotype via EMT after injury, which is a potential source of profibrotic cytokines and ECM deposition, thus leading to gradual aggravation of renal fibrosis (Qi & Yang, 2018).

    EMT is the process by which a polarized epithelial cell adopts a mesenchymal cell phenotype, and includes enhanced migratory ability, invasiveness, resistance to apoptosis, and secretion of ECM (Kalluri & Weinberg, 2009).EMT is manifested by a decrease in epithelial markers, such as E-cadherin and zonula occludens-1 (ZO-1), and the acquisition of mesenchymal proteins, including vimentin, α-SMA, fibronectin, and collagen I (Liu, 2010). Anorga et al.(2018) reported that renal injury can result in elevated TGF-β1 expression, leading to TAZ protein induction and nuclear accumulation. Stable TAZ overexpression in RTECs promotes fibrotic factor expression (including CTGF, fibronectin, and plasminogen activator inhibitor (PAI)-1)), dedifferentiation (as evident by altered morphology, increased mesenchymal marker expression, and loss of epithelial characteristics), and growth inhibition via Smad3-dependent CTGF up-regulation.Seo et al. (2016) found that TEC-specific deletion of Sav1 can increase TAZ protein expression and YAP1 nuclear localization after UUO, resulting in TGF-β and TGF-β receptor II up-regulation and aberrant activation of Wnt/β-catenin signaling, thereby aggravating renal TIF and enhancing EMTlike phenotype changes. Sav1 loss can also induce Stat3 activation and SASP with the accumulation of collagen and vimentin, leading to a fibrosis phenotype that can be further enhanced by AKI with aristolochic acid (AA) (Leung et al.,2017). Treatment with VP (YAP inhibitor) inhibits the activation of genes associated with senescence, SASP, and Stat3, and impedes the development of fibrosis (Leung et al., 2017).Therefore, inhibition of the Hippo pathway may mediate the occurrence of EMT, and YAP/TAZ may serve as a new therapeutic target for chronic renal fibrosis progress after AKI.

    Crosstalk between Hippo pathway and fibroblasts/myofibroblasts

    Myofibroblasts are important in ECM production during TIF.Myofibroblasts are rare in normal kidneys but markedly increase during chronic fibrosis, with possible precursors including interstitial stromal cells, such as fibroblasts,pericytes, and mesenchymal stem cells (MSC), as well as bone marrow-derived cells and endothelial cells. Current evidence suggests that stromal cells are the major origin of myofibroblasts (Iwano et al., 2002; LeBleu et al., 2013; Yuan et al., 2019). Whether epithelial cells can turn into myofibroblasts has been debated for many years. However,recent studies suggest that epithelial cells contribute little to the myofibroblast population after AKI (Humphreys et al.,2010; Koesters et al., 2010; LeBleu et al., 2013).Myofibroblasts are the major source of ECM proteins, crosslinking enzymes, and inhibitors of matrix-degrading metalloproteinases, characterized by the expression of the classic marker α-SMA (Chen et al., 2020; Gewin, 2018).

    Upon insult, maladaptive RTECs cause a switch in the microenvironment of the tubulointerstitial area where myofibroblasts are activated (Qi & Yang, 2018). Resident fibroblasts proliferate and transition into myofibroblasts by profibrotic factors, such as CTGF, which are synthesized and released from injured RPTCs through a YAP activationdependent pathway (Chen et al., 2020). Szeto et al. (2016)suggested that soft ECM inhibits while stiff ECM augments TGF-β-induced Smad2/3 nuclear accumulation mediated by YAP/TAZ, leading to fibroblast-myofibroblast transition and renal fibrosis. Furthermore, treating cells and mice with VP can reduce YAP/TAZ levels and block TGF-β-induced myofibroblast activation. TGF-β1 can also up-regulate YAP/TAZ expression through mTORC2 signaling-stimulated YAP/TAZ transcriptional activation, thereby stimulating fibroblast activation (Gui et al., 2018). Liang et al. (2017)demonstrated that fibroblasts cultured on a stiff matrix transform into myofibroblasts in a process dependent on YAP activation, even in the absence of TGF-β. Furthermore, Gli1+cell-specific knockout of YAP/TAZ in mice suppresses UUOinduced ECM deposition, myofibroblast accumulation, and TIF. Therefore, YAP acts as a tissue mechanosensor in the kidney and can be activated by ECM to transform fibroblasts into myofibroblasts. The interaction between YAP/TAZ and ECM forms a positive feedback loop, resulting in kidney fibrosis.

    Correlation between Hippo pathway and G2/M cell cycle arrest in renal fibrosis

    Cell cycle arrest in RTECs during maladaptive repair is an important driver of kidney fibrogenesis (Yang et al., 2011). In uninjured kidneys, most RTECs are quiescent and remain in the G0 phase of the cell cycle. Upon injury, cells may enter the cell cycle to replace cells lost during the insult, with some cells arrested in either the G1 or G2 phase to allow time for DNA damage repair (Gewin, 2018). Accumulation of cells arrested in the G2/M growth phase is a common feature of progressive fibrotic kidney disease, resulting in the acquisition of a pathogenic phenotype characterized by sustained synthesis and secretion of profibrotic factors (Bonventre, 2014;Ferenbach & Bonventre, 2015).

    As YAP/TAZ are involved in cell proliferation and survival,Yap activation can promote cell cycle progression (Chen et al.,2018; Mizuno et al., 2012). Thus, whether YAP/TAZ are associated with G2/M phase cell cycle arrest warrants investigation. Anorga et al. (2018) found that TAZoverexpressing epithelial cells exhibit epithelial growth inhibition and G2/M cell cycle arrest after injury, consistent with the enhanced expression of growth-arrest genep21,conferring a fibrotic phenotype. Down-regulation of YAP in human laryngeal carcinoma Hep-2 cells induces G2/M cell cycle arrest, reduces G0/G1 phase percentage, and increases apoptosis, indicating that YAP inactivation is related to G2/M arrest (Tang et al., 2019). However, other studies have shown that tubular maladaptive repair in the kidney is associated with YAP activation and that up-regulation of YAP induces an increase in the G2/M- and S-phases and a decrease in the G0/G1-phase in HK-2 cells, thus mediating kidney fibrosis after acute injury (Xu et al., 2016; Zheng et al., 2021). Clearly,these two hypotheses conflict with each other and there may exist a feedback regulation against G2/M phase arrest by triggering YAP activation in the kidney. Furthermore, Gerhardt et al. (2021) suggested the Hippo is involved in early injured RPTCs in mice after moderate AKI based on up-regulation of TEAD1 regulon. They also reported up-regulation of regulators involved in YAP-inactivation and Hippo signaling (e.g.,Sav1,Stk3), as well as effectors of the transcriptional response after Hippo silencing (e.g.,Yap1,Tead 1) and YAP-target genes such asAxlandCtgf. In particular, they identifiedVcam1+/Ccl2+ RPTCs at a late injury stage with marked activation of NF-κB-, TNF-, and AP-1-signaling pathways. This population of RPTCs showed features of SASP but did not exhibit G2/M cell cycle arrest, distinct from other reports of maladaptive RPTCs following kidney injury. However, the exact associations between YAP/TAZ and G2/M cell cycle arrest and whether G2/M cell cycle arrest exists in RPTCs that fail to repair after injury require further investigation.

    Function of Hippo pathway in macrophage and inflammation regulation

    AKI is typically accompanied by immune activation and inflammation, which involve macrophage infiltration. Recruited macrophages exert versatile effects on kidney repair after injury depending on their polarization form: i.e.,proinflammatory M1 phenotype and reparative M2 phenotype.The predominant M1 phenotype in the early phase of AKI produces proinflammatory cytokines and mediators, which, in turn, exacerbate inflammation (Ferenbach & Bonventre, 2015;Qi & Yang, 2018). The M2 polarization of macrophages in the resolution phase of AKI is beneficial for renal recovery.However, the M2 phenotype may exert a pro-fibrotic influence by promoting TIF, expressing matrix metalloproteases, and secreting pro-fibrotic factors (Gewin et al., 2017). Therefore,persistent activation of M2 macrophages is instrumental in maladaptive repair and fibrosis in CKD (Baek, 2019).

    Hippo-YAP pathway activation after ischemic AKI is associated with tubular maladaptive repair and interstitial macrophage infiltration mediated by MCP-1 up-regulation,while inhibition of YAP nuclear translocation by VP attenuates macrophage infiltration and TIF (Zheng et al., 2021). VP treatment in UUO-induced AKI mice also decreases macrophage recruitment and cell adhesion molecule expression (Jin et al., 2020). Tubular cell-specific knockout of MST1/2 promotes the expression of inflammatory factors and macrophage marker F4/80; however, expression returns to physiological levels after YAP deletion in MST1/2 double knockout mice (Xu et al., 2020a). These data suggest that YAP may play a pivotal role in chronic inflammation during maladaptive repair after renal injury.

    Feng et al. (2018b) demonstrated that YAP/TAZ induction mediates Wnt5a and TGF-β1-promoted macrophage M2 polarization and contributes to renal fibrosis after UUO or I/R injury in mice, whereas specific ablation of the TAZ gene in macrophages can markedly decrease the M2 phenotype and kidney fibrosis. However, further studies are needed to decipher the exact mechanism by which YAP/TAZ regulate macrophage M2 polarization.

    Hippo-YAP pathway activation can lead to renal hypertrophy through RTEC enlargement

    Renal hypertrophy is characterized by increased organ size due to cell enlargement rather than cell proliferation (Fine &Norman, 1989). In the kidney, the growth of remaining renal tissue in response to renal tissue loss is known as renal compensatory hypertrophy (RCH) and can help restore normal kidney function following disease or loss of kidney tissue(Sugaya et al., 2000). As the proximal tubule forms the bulk of kidney mass, this nephron segment contributes the most to renal hypertrophy (Hayslett et al., 1968).

    In renal epithelial cells, increased nuclear accumulation and transcriptional activity of YAP can trigger activation of the Akt/mTOR signaling pathway and its downstream target S6K1.This leads to silencing of the peripheral tight junction protein zona occludens 2 (ZO-2), which induces an increase in cell size and a higher protein/DNA ratio, thereby triggering RCH(Domínguez-Calderón et al., 2016). ZO-2 is also a positive regulator of the Hippo pathway as it associates with LATS1 and functions as a pathway scaffold (González-González et al., 2021). However, chronic cell expansion beyond optimal conditions may present certain disadvantages, and hemodynamic and metabolic hyperfunction of residual nephrons may prove maladaptive over time (Lafferty &Brenner, 1990; Nath, 1992). Excessive RCH can lead to tubular atrophy, interstitial fibrosis, and progressive decline in renal function (Hostetter, 1995). Therefore, persistent YAP activation after AKI may also cause renal hypertrophy and accelerate progression towards CKD (Figure 4).

    POTENTlAL ROLE OF HlPPO PATHWAY lN FERROPTOSlS AFTER AKl

    Ferroptosis is characterized by intracellular iron accumulation and lipid peroxidation during cell death (Dixon et al., 2012).The characteristics of ferroptosis differ from necrosis,apoptosis, and autophagy in cell morphology and function (Xie et al., 2016). The morphology of ferroptotic cells includes mitochondrial changes, such as reduced volume, decreased mitochondrial cristae, and increased membrane density(Wang et al., 2021). However, the nucleus remains normal,with no chromatin agglutination (Wang et al., 2020a).Following ferroptosis, intracellular glutathione (GSH)consumption and glutathione peroxidase 4 (GPX4) inactivation occur (Li et al., 2020). Recent studies have shown that ferroptosis participates in various pathological models of AKI and classical ferroptosis inhibitors, such as ferrostatin-1 and liproxstatin-1, are capable of interfering with key molecules in the ferroptosis signaling pathway to resist AKI (Hu et al., 2019;Martin-Sanchez et al., 2017; Skouta et al., 2014; Zilka et al.,2017). GPX4 deletion in mice can spontaneously trigger AKI(Friedmann Angeli et al., 2014), whereas GPX4 up-regulation prevents AKI (Zhang et al., 2021).

    A growing number of studies have revealed the relationship between the Hippo pathway and ferroptosis in cancer cells.Yang & Chi (2020) suggested that triggering ferroptosis may have therapeutic potential for TAZ-activated tumors.Ferroptosis sensitivity in renal and ovarian cells is regulated by cell density through the TAZ-EMP-NOX4 and TAZANGPTL4-NOX2 pathways, respectively (Yang et al., 2019,2020). TAZ removal confers ferroptosis resistance, while overexpression of the constitutively active form of TAZ(TAZS89A) sensitizes cells to ferroptosis. YAP exerts a similar function in cancer cell ferroptosis, with knockdown of YAP or its direct target gene S-phase kinase-associated protein 2(SKP2) shown to robustly protect cells and abolish lipid peroxidation during erastin-induced ferroptosis (Yang et al.,2021). However, despite current evidence supporting the connection between Hippo pathway effectors and ferroptosis in cancer, there is a lack of research on the role of the Hippo pathway in ferroptosis during AKI progression. Moreover, the specific role of ferroptosis in AKI caused by different etiologies remains unclear. Thus, further exploration in this field is necessary for the clinical treatment of AKI.

    HlPPO PATHWAY MAY CONTRlBUTE TO KlDNEY CANCER DEVELOPMENT lN AKl

    Renal cell carcinoma (RCC), which is derived from RTECs,accounts for 85% of all renal malignancies (Cinar et al., 2021;Kovacs et al., 1997). Emerging evidence suggests that dysregulation of Hippo-YAP1 signaling plays a significant role in the etiology of aggressive kidney cancer (Han, 2019). YAP1 protein accumulates in the nuclei of clear cell RCC (ccRCC)cells, although normal kidney cells primarily express the cytoplasmic YAP1 protein (Godlewski et al., 2018). Similarly,nuclear YAP1 is considerably higher in ccRCC cells than in unaltered proximal kidney cortices (Rybarczyk et al., 2017).Up-regulation of YAP1 in ccRCC patients is also associated with poorer clinical outcomes (Chen et al., 2014). Thus,nuclear YAP1 appears to play an oncogenic role in ccRCC cells, promoting cell proliferation and survival (Rybarczyk et al., 2017).

    Recent studies have identified AKI as a risk factor for renal cancer development (Kusmartsev, 2021). Based on data collected from several multicenter studies, Peired et al. (2020)found that AKI can significantly increase the risk of papillary RCC (pRCC) development and tumor relapse in humans.They also induced I/R injury in wild-type mice and examined their kidneys at different timepoints and found that hypoxia promotes the undifferentiated cell state in various stem and precursor populations by activating Notch-responsive promoters and increasing expression of genes downstream of Notch, driving a hypoxia-inducible factor HIF1a-to-HIF2a switch favoring transformation into cancer stem cells and supporting tumor growth. Notably, YAP/TAZ activity is linked to oxygen availability (Yu et al., 2015). Under hypoxic conditions, HIF1 stimulates the expression of SIAH1/2, two E3 ubiquitin ligases. SIAH1/2 then promote ubiquitination and degradation of LATS2, leading to YAP/TAZ activation (Ma et al., 2015; Xiang et al., 2014). In addition, HIF1 directly induces the transcription of TAZ (Xiang et al., 2014), and YAP interacts with and stabilizes HIF1 to enhance the transcription of HIF1 target genes (Ma et al., 2015). Furthermore, there are two main modalities of YAP/TAZ-Notch signaling crosstalk: i.e.,YAP/TAZ-mediated transcriptional regulation of Notch ligands or receptors and transcriptional co-regulation of common direct target genes by YAP/TAZ and the Notch intracellular domain (NICD) (Totaro et al., 2018). Whether the Hippo pathway exerts a function in pRCC development after AKI deserves further clarification.

    Zhou et al. (2021) also investigated the relationship between AKI-associated systemic inflammation and risk of kidney cancer development. They subjected genetically modified mice with Cre-mediated deletion of theTrp53andPtengenes in Ggt1-expressing RPTCs (GPPY mice) to I/R injury to induce acute injury in one kidney. Their results showed that I/R injury-induced AKI promoted the formation of ccRCC in the contralateral uninjured kidney, but not in the kidney subjected to I/R injury. A significant increase in tissue infiltration of neutrophils and fibroblasts was also observed,associated with the up-regulation of inflammatory factors CXCL1, CXCL13, TIMP1, and ILRα, with CXCL1 displaying the highest levels of up-regulation locally and systemically.Notably, CXCL1 expression was predominantly associated with dysplastic epithelial cells in the I/R-injured kidney, but not in the contralateral uninjured kidney. Up-regulation of CXCR2,which serves as a natural receptor for CXCL1, was also observed in stromal fibroblasts, mostly in the uninjured kidney.Administration of anti-CXCR2 antibodies blocked CXCL1/CXCR2 signaling, leading to a reduction in the recruitment of neutrophils, fibroblasts, and most importantly, RCC incidence.Chen et al. (2018) also demonstrated an increase in YAP expression and YAP-TEAD interaction in RPTCs from post-I/R-injured mouse kidneys. Activation of TEAD1 can lead to an increase in the expression of its target, CXCL1, i.e.,chemokine-mediated neutrophil recruitment (Kusumanchi et al., 2021). Zheng et al. (2021) also proved that renal CXCL2 and CXCR2 are markedly increased in post-AKI mice through tubular YAP activation. Hence, growing evidence suggests that the Hippo pathway is involved in the positive association between ccRCC formation and AKI-induced systemic inflammation.

    CONCLUSlONS AND FUTURE PERSPECTlVES

    Research published over the last several years has revealed the significant role that the Hippo signaling pathway plays in pathological changes of AKI and subsequent repair. During renal pathological injury in AKI, MST1, the upstream factor of the Hippo pathway, mediates RTEC apoptosis by inducing mitochondrial fission, thereby aggravating tubular damage.Transient overexpression of YAP and its downstream mediatorsurvivinin the aftermath of AKI can result in acquired cytoresistance in RPTCs to protect the kidney against further ischemia or nephrotoxicity. Moreover, the Hippo pathway elicits both beneficial and detrimental effects on recovery after AKI. If the injury is mild/moderate, downstream factors YAP/TAZ can be transiently activated, leading to complete/adaptive renal repair. However, if the injury is severe and recurring, the constant increase and activation of YAP/TAZ can lead to incomplete/maladaptive renal repair and progressive fibrotic CKD via several mechanisms, including EMT, fibroblast to myofibroblast conversion, G2/M phase cell cycle arrest, macrophage infiltration, and renal hypertrophy.Although ferroptosis is implicated in various pathological models of AKI and Hippo pathway effectors are highly associated with ferroptosis in cancer cells, whether the Hippo pathway is involved in ferroptosis in AKI progression requires further investigation. In addition, while AKI is a risk factor of kidney cancer development, including ccRCC and pRCC,whether the Hippo pathway participates in this process remains to be clarified. Hence, despite compelling evidence,the correlation between the Hippo pathway and kidney injury still requires additional research, and the potential mechanisms and signaling pathways need to be further elucidated. Furthermore, pharmacological manipulation of the Hippo pathway for the treatment of AKI is uncertain and requires future exploration.

    COMPETlNG lNTERESTS

    The authors declare that they have no competing interests.

    AUTHORS’ CONTRlBUTlONS

    C.Z., C.L.L., K.X.X., Z.H.Z., and G.Z.C. conceived and designed this review article. C.Z. wrote the initial draft of the manuscript and drew the figures. J.L. and H.J.W. reviewed and edited the manuscript. All authors read and approved the final version of the manuscript.

    又黄又粗又硬又大视频| 我要搜黄色片| 女人被狂操c到高潮| 黄色 视频免费看| 1024手机看黄色片| 韩国av一区二区三区四区| 亚洲九九香蕉| 国产精品一区二区精品视频观看| 午夜精品久久久久久毛片777| 成在线人永久免费视频| 日本免费一区二区三区高清不卡| 国内精品一区二区在线观看| 国产精品野战在线观看| 免费在线观看成人毛片| 99在线人妻在线中文字幕| 好男人电影高清在线观看| a级毛片a级免费在线| 国产久久久一区二区三区| 国产不卡一卡二| 亚洲色图 男人天堂 中文字幕| 男人舔女人的私密视频| 最新在线观看一区二区三区| 国产一级毛片七仙女欲春2| avwww免费| 精品无人区乱码1区二区| 国产精品99久久99久久久不卡| 国产精品 国内视频| 亚洲电影在线观看av| 国产精品久久电影中文字幕| 国产精品香港三级国产av潘金莲| 亚洲第一欧美日韩一区二区三区| 在线永久观看黄色视频| 一级毛片精品| 午夜免费观看网址| 中文资源天堂在线| 老鸭窝网址在线观看| 国产三级中文精品| 精品久久久久久久末码| 麻豆久久精品国产亚洲av| 国产区一区二久久| 亚洲熟妇中文字幕五十中出| 欧美高清成人免费视频www| 97超级碰碰碰精品色视频在线观看| 精品国产乱码久久久久久男人| 2021天堂中文幕一二区在线观| 最近最新中文字幕大全电影3| 99riav亚洲国产免费| 亚洲五月天丁香| 日日干狠狠操夜夜爽| 久久久久久国产a免费观看| 黄色 视频免费看| 国产精品免费视频内射| 精品国内亚洲2022精品成人| svipshipincom国产片| 色综合欧美亚洲国产小说| 18美女黄网站色大片免费观看| 国产黄a三级三级三级人| 精品一区二区三区视频在线观看免费| 久久午夜综合久久蜜桃| 久久久久国内视频| 国产精品香港三级国产av潘金莲| 亚洲中文字幕一区二区三区有码在线看 | 国产欧美日韩精品亚洲av| 日本成人三级电影网站| 亚洲精品av麻豆狂野| 国产片内射在线| 国产视频内射| 99在线视频只有这里精品首页| 少妇裸体淫交视频免费看高清 | 久久婷婷人人爽人人干人人爱| 成人国产一区最新在线观看| 亚洲午夜理论影院| 亚洲国产精品久久男人天堂| 美女 人体艺术 gogo| 久久久久久国产a免费观看| 色在线成人网| 亚洲欧美精品综合一区二区三区| 一本精品99久久精品77| 久久天躁狠狠躁夜夜2o2o| 日本精品一区二区三区蜜桃| 国产爱豆传媒在线观看 | 老汉色av国产亚洲站长工具| 女人高潮潮喷娇喘18禁视频| 中文亚洲av片在线观看爽| 亚洲av中文字字幕乱码综合| x7x7x7水蜜桃| 亚洲全国av大片| 欧美日韩瑟瑟在线播放| 日本在线视频免费播放| 国产精品久久久久久人妻精品电影| 国内毛片毛片毛片毛片毛片| 免费在线观看视频国产中文字幕亚洲| 亚洲成a人片在线一区二区| 久久精品国产亚洲av高清一级| www.999成人在线观看| 在线观看www视频免费| 超碰成人久久| 波多野结衣巨乳人妻| 制服人妻中文乱码| 婷婷丁香在线五月| 午夜激情福利司机影院| a在线观看视频网站| 亚洲无线在线观看| 在线免费观看的www视频| 国产精品98久久久久久宅男小说| 淫妇啪啪啪对白视频| 免费看日本二区| 亚洲人与动物交配视频| 成在线人永久免费视频| 亚洲av成人av| av欧美777| 人妻夜夜爽99麻豆av| av福利片在线观看| 一本综合久久免费| 亚洲人成网站高清观看| 精品欧美一区二区三区在线| 欧美成狂野欧美在线观看| 欧美日本亚洲视频在线播放| 夜夜看夜夜爽夜夜摸| 国产伦在线观看视频一区| 校园春色视频在线观看| 日韩有码中文字幕| 日本 欧美在线| 国产精品日韩av在线免费观看| 国产主播在线观看一区二区| 国产精品久久久久久亚洲av鲁大| 亚洲成人免费电影在线观看| 日韩国内少妇激情av| 国产97色在线日韩免费| 黄片大片在线免费观看| 欧美成人性av电影在线观看| 国产高清激情床上av| 欧美+亚洲+日韩+国产| 在线观看免费视频日本深夜| 亚洲一区二区三区不卡视频| 国产在线精品亚洲第一网站| 久久伊人香网站| av片东京热男人的天堂| 一级作爱视频免费观看| 亚洲人成电影免费在线| 日韩三级视频一区二区三区| 亚洲精品久久成人aⅴ小说| 18禁观看日本| 精品少妇一区二区三区视频日本电影| 中文字幕高清在线视频| 午夜成年电影在线免费观看| 欧美成狂野欧美在线观看| 国产精品 国内视频| 亚洲熟妇熟女久久| 欧美午夜高清在线| 一级a爱片免费观看的视频| 老汉色∧v一级毛片| 国产精品一区二区三区四区免费观看 | 亚洲真实伦在线观看| 日本黄色视频三级网站网址| 国产精品一区二区三区四区免费观看 | 青草久久国产| 日韩三级视频一区二区三区| 国产精品免费一区二区三区在线| 伊人久久大香线蕉亚洲五| 国内少妇人妻偷人精品xxx网站 | 一本精品99久久精品77| 亚洲av电影不卡..在线观看| 亚洲avbb在线观看| www国产在线视频色| 国产日本99.免费观看| 成人av在线播放网站| 国产精品 国内视频| 夜夜爽天天搞| 欧美乱妇无乱码| 黄色视频不卡| 88av欧美| 宅男免费午夜| 免费电影在线观看免费观看| 中文字幕人成人乱码亚洲影| 欧美av亚洲av综合av国产av| 亚洲精品在线观看二区| 身体一侧抽搐| 熟女电影av网| 在线观看免费视频日本深夜| 曰老女人黄片| 中文资源天堂在线| 99热这里只有精品一区 | 男女午夜视频在线观看| www日本在线高清视频| 波多野结衣巨乳人妻| 国产精品野战在线观看| 欧美最黄视频在线播放免费| 国产精品一区二区免费欧美| 两个人看的免费小视频| 精品久久久久久久久久久久久| 国产激情久久老熟女| 亚洲免费av在线视频| 在线观看免费午夜福利视频| 亚洲欧美日韩高清专用| 一本一本综合久久| 亚洲一码二码三码区别大吗| 在线观看美女被高潮喷水网站 | 无限看片的www在线观看| 女人爽到高潮嗷嗷叫在线视频| 黄色毛片三级朝国网站| 亚洲欧美日韩高清在线视频| 99久久99久久久精品蜜桃| 精品国产乱子伦一区二区三区| 中文在线观看免费www的网站 | 国产三级中文精品| 久久婷婷成人综合色麻豆| 在线观看www视频免费| 亚洲成人久久性| 精品电影一区二区在线| 日韩欧美 国产精品| 国产午夜福利久久久久久| 国产一区二区三区视频了| 每晚都被弄得嗷嗷叫到高潮| 亚洲真实伦在线观看| 国产在线观看jvid| 身体一侧抽搐| 黄色 视频免费看| 国产爱豆传媒在线观看 | 美女黄网站色视频| 精品国产美女av久久久久小说| 中文字幕人成人乱码亚洲影| 精华霜和精华液先用哪个| 少妇人妻一区二区三区视频| 亚洲国产看品久久| 黄色女人牲交| 亚洲精品美女久久久久99蜜臀| 老汉色av国产亚洲站长工具| 久久久久久久午夜电影| 黄片小视频在线播放| 他把我摸到了高潮在线观看| 久久精品aⅴ一区二区三区四区| 国产成人系列免费观看| 精品电影一区二区在线| av天堂在线播放| 国产在线观看jvid| 一进一出好大好爽视频| 最近视频中文字幕2019在线8| 五月玫瑰六月丁香| 天堂动漫精品| 精品欧美一区二区三区在线| 久久国产精品人妻蜜桃| 在线国产一区二区在线| 中文资源天堂在线| 黄色片一级片一级黄色片| 18美女黄网站色大片免费观看| 久久中文字幕一级| 91av网站免费观看| 日本a在线网址| 香蕉丝袜av| av超薄肉色丝袜交足视频| 久久亚洲真实| 免费看十八禁软件| 日韩大码丰满熟妇| 麻豆av在线久日| 色精品久久人妻99蜜桃| 免费看十八禁软件| 真人一进一出gif抽搐免费| 欧美日韩福利视频一区二区| 无人区码免费观看不卡| 欧美丝袜亚洲另类 | 国产又黄又爽又无遮挡在线| 最新美女视频免费是黄的| 久久人妻av系列| 日日爽夜夜爽网站| 搡老熟女国产l中国老女人| 岛国在线观看网站| 一级黄色大片毛片| 国产99白浆流出| 每晚都被弄得嗷嗷叫到高潮| 一区二区三区高清视频在线| 麻豆久久精品国产亚洲av| 天天躁夜夜躁狠狠躁躁| 欧美日本亚洲视频在线播放| 日韩精品免费视频一区二区三区| 国产免费av片在线观看野外av| 1024香蕉在线观看| 欧美黄色片欧美黄色片| 99精品欧美一区二区三区四区| 床上黄色一级片| 久久久国产精品麻豆| 国产亚洲精品久久久久5区| 日韩av在线大香蕉| 免费看十八禁软件| 99riav亚洲国产免费| 欧美成人一区二区免费高清观看 | 久久精品国产亚洲av高清一级| 1024视频免费在线观看| 别揉我奶头~嗯~啊~动态视频| 中文字幕精品亚洲无线码一区| 狂野欧美激情性xxxx| 伦理电影免费视频| 精品一区二区三区av网在线观看| 啦啦啦韩国在线观看视频| 黑人操中国人逼视频| 白带黄色成豆腐渣| 国产一级毛片七仙女欲春2| 99国产极品粉嫩在线观看| 一个人免费在线观看电影 | 日本 欧美在线| 色尼玛亚洲综合影院| 精品不卡国产一区二区三区| 久久久国产欧美日韩av| 天天添夜夜摸| 国产单亲对白刺激| 在线观看66精品国产| 亚洲熟妇中文字幕五十中出| 日韩有码中文字幕| 久久久久免费精品人妻一区二区| 国产成人系列免费观看| 久久久久久久精品吃奶| 最新美女视频免费是黄的| 12—13女人毛片做爰片一| 舔av片在线| 很黄的视频免费| 日韩欧美在线乱码| 久久精品亚洲精品国产色婷小说| 男人舔女人的私密视频| 露出奶头的视频| 午夜激情福利司机影院| 波多野结衣巨乳人妻| 国产精品乱码一区二三区的特点| 我的老师免费观看完整版| 美女大奶头视频| 十八禁网站免费在线| 国产成人精品久久二区二区91| 久久久国产成人免费| 狂野欧美激情性xxxx| 啦啦啦观看免费观看视频高清| 免费在线观看视频国产中文字幕亚洲| 午夜两性在线视频| 少妇人妻一区二区三区视频| 国产91精品成人一区二区三区| 一二三四社区在线视频社区8| 亚洲中文av在线| 狠狠狠狠99中文字幕| 成人手机av| 欧美一级毛片孕妇| 成人18禁高潮啪啪吃奶动态图| 黑人操中国人逼视频| 在线观看66精品国产| 国产男靠女视频免费网站| 久久精品国产亚洲av香蕉五月| 国产一区二区激情短视频| 一个人免费在线观看电影 | 琪琪午夜伦伦电影理论片6080| 搡老妇女老女人老熟妇| 亚洲色图 男人天堂 中文字幕| 色av中文字幕| 99国产精品99久久久久| 国产精品1区2区在线观看.| av天堂在线播放| 中文字幕人妻丝袜一区二区| 免费看美女性在线毛片视频| 久久九九热精品免费| 可以免费在线观看a视频的电影网站| 日本一区二区免费在线视频| 免费在线观看黄色视频的| 特大巨黑吊av在线直播| 中文字幕精品亚洲无线码一区| 色综合欧美亚洲国产小说| 很黄的视频免费| 观看免费一级毛片| 又黄又粗又硬又大视频| 在线观看www视频免费| av在线播放免费不卡| 久久精品综合一区二区三区| 麻豆av在线久日| 国产亚洲欧美98| 国产99久久九九免费精品| 国产精品1区2区在线观看.| 村上凉子中文字幕在线| 国产av不卡久久| 成人午夜高清在线视频| 99久久国产精品久久久| 99久久综合精品五月天人人| 看免费av毛片| 欧美日韩国产亚洲二区| 国语自产精品视频在线第100页| 欧美+亚洲+日韩+国产| 精品乱码久久久久久99久播| 国产一区二区在线av高清观看| 天天添夜夜摸| 女警被强在线播放| 亚洲在线自拍视频| 黄色成人免费大全| 欧美在线黄色| 成在线人永久免费视频| 国产精品亚洲av一区麻豆| 国产精品一区二区三区四区免费观看 | 色精品久久人妻99蜜桃| 十八禁人妻一区二区| 欧美黄色淫秽网站| www.熟女人妻精品国产| 国产亚洲精品一区二区www| 国产精品综合久久久久久久免费| 丁香欧美五月| 国产亚洲欧美在线一区二区| 黄片大片在线免费观看| 亚洲男人的天堂狠狠| 国产亚洲精品av在线| 99久久久亚洲精品蜜臀av| 男人舔女人下体高潮全视频| 日本免费a在线| 精华霜和精华液先用哪个| 成人三级做爰电影| 国产精品99久久99久久久不卡| 亚洲中文av在线| 免费在线观看亚洲国产| 欧美 亚洲 国产 日韩一| 99精品欧美一区二区三区四区| 美女 人体艺术 gogo| 老司机在亚洲福利影院| av有码第一页| 91麻豆av在线| 精品少妇一区二区三区视频日本电影| 亚洲熟妇熟女久久| 狂野欧美白嫩少妇大欣赏| 最近最新中文字幕大全电影3| 一进一出抽搐gif免费好疼| 国产精品日韩av在线免费观看| tocl精华| 九色国产91popny在线| 成人av一区二区三区在线看| 亚洲人成电影免费在线| x7x7x7水蜜桃| 黄色a级毛片大全视频| 亚洲精品一卡2卡三卡4卡5卡| 亚洲乱码一区二区免费版| 亚洲欧美日韩无卡精品| 免费观看人在逋| 老司机午夜十八禁免费视频| 国产精品国产高清国产av| 一a级毛片在线观看| 日本 欧美在线| 色av中文字幕| 免费在线观看视频国产中文字幕亚洲| av中文乱码字幕在线| 国产精品爽爽va在线观看网站| 狠狠狠狠99中文字幕| 国产成人av教育| 久久伊人香网站| 在线a可以看的网站| 国产精品免费一区二区三区在线| 女警被强在线播放| 亚洲va日本ⅴa欧美va伊人久久| 久久天堂一区二区三区四区| 黄色片一级片一级黄色片| 亚洲最大成人中文| 长腿黑丝高跟| 色av中文字幕| 一进一出抽搐gif免费好疼| 狠狠狠狠99中文字幕| 中文字幕久久专区| 成人三级黄色视频| 国产亚洲av嫩草精品影院| 国产亚洲精品一区二区www| 黄片小视频在线播放| 老司机午夜十八禁免费视频| 一二三四在线观看免费中文在| 久久久久久国产a免费观看| 免费在线观看成人毛片| 色老头精品视频在线观看| 男女床上黄色一级片免费看| 欧美黄色淫秽网站| 欧美激情久久久久久爽电影| 十八禁网站免费在线| 色av中文字幕| 日韩精品青青久久久久久| 国产精品 国内视频| 韩国av一区二区三区四区| 在线永久观看黄色视频| 亚洲欧美激情综合另类| 国产亚洲av高清不卡| 亚洲性夜色夜夜综合| 亚洲av成人精品一区久久| av在线播放免费不卡| 国产精品美女特级片免费视频播放器 | 香蕉丝袜av| 久久久久久久久免费视频了| 国产av在哪里看| 一本久久中文字幕| 亚洲va日本ⅴa欧美va伊人久久| 亚洲精品色激情综合| 久久草成人影院| 亚洲成人久久性| 中亚洲国语对白在线视频| 真人做人爱边吃奶动态| 国产蜜桃级精品一区二区三区| 国内精品久久久久久久电影| 国产视频一区二区在线看| 天天添夜夜摸| 一级毛片女人18水好多| 国内揄拍国产精品人妻在线| 精品一区二区三区四区五区乱码| 亚洲国产高清在线一区二区三| 最近视频中文字幕2019在线8| 黑人欧美特级aaaaaa片| 一级a爱片免费观看的视频| 一进一出抽搐动态| 欧美精品亚洲一区二区| 露出奶头的视频| 黄色丝袜av网址大全| 国产在线观看jvid| 美女 人体艺术 gogo| 国产午夜福利久久久久久| 欧美成人性av电影在线观看| 一区二区三区国产精品乱码| 亚洲精品色激情综合| 欧美一级毛片孕妇| 国产爱豆传媒在线观看 | 中文资源天堂在线| 男女床上黄色一级片免费看| 国产精品永久免费网站| 人人妻人人澡欧美一区二区| 最近在线观看免费完整版| 国产主播在线观看一区二区| 很黄的视频免费| 老汉色∧v一级毛片| 精品久久久久久久末码| 一区二区三区国产精品乱码| 十八禁网站免费在线| 亚洲人成网站高清观看| 久久精品aⅴ一区二区三区四区| 国产成人aa在线观看| 日本黄色视频三级网站网址| 欧美成人性av电影在线观看| 久久精品亚洲精品国产色婷小说| 男女床上黄色一级片免费看| 欧美大码av| 啦啦啦免费观看视频1| 欧美色欧美亚洲另类二区| 不卡一级毛片| 国产精华一区二区三区| 国产黄片美女视频| 国产亚洲精品久久久久5区| 欧美最黄视频在线播放免费| 欧美午夜高清在线| 亚洲一区高清亚洲精品| 人人妻,人人澡人人爽秒播| 亚洲人与动物交配视频| 动漫黄色视频在线观看| 久久精品91无色码中文字幕| 超碰成人久久| 丝袜美腿诱惑在线| 人人妻人人看人人澡| 五月玫瑰六月丁香| 精品久久久久久成人av| 亚洲乱码一区二区免费版| 99久久精品国产亚洲精品| 亚洲专区中文字幕在线| 亚洲欧美日韩高清专用| 国语自产精品视频在线第100页| 中文字幕熟女人妻在线| 亚洲av中文字字幕乱码综合| 欧美黑人欧美精品刺激| 91大片在线观看| 91麻豆精品激情在线观看国产| 日本撒尿小便嘘嘘汇集6| 99久久久亚洲精品蜜臀av| 俺也久久电影网| 中文亚洲av片在线观看爽| 这个男人来自地球电影免费观看| 亚洲国产精品合色在线| 免费看美女性在线毛片视频| 成人永久免费在线观看视频| 久久久久久国产a免费观看| 亚洲 欧美一区二区三区| 久久99热这里只有精品18| aaaaa片日本免费| 亚洲 国产 在线| 欧美人与性动交α欧美精品济南到| 熟女电影av网| cao死你这个sao货| 国内精品一区二区在线观看| 淫秽高清视频在线观看| 免费一级毛片在线播放高清视频| 天堂√8在线中文| 天堂影院成人在线观看| 少妇被粗大的猛进出69影院| 亚洲av电影不卡..在线观看| 午夜福利欧美成人| 在线观看免费午夜福利视频| ponron亚洲| 99热6这里只有精品| 国产精品综合久久久久久久免费| 精品欧美国产一区二区三| 日韩中文字幕欧美一区二区| 嫁个100分男人电影在线观看| 麻豆成人av在线观看| 欧美成人一区二区免费高清观看 | 日日爽夜夜爽网站| АⅤ资源中文在线天堂| 可以在线观看毛片的网站| 亚洲国产欧美人成| 啦啦啦韩国在线观看视频| 国产成年人精品一区二区| 亚洲在线自拍视频| 国产欧美日韩精品亚洲av| 欧美最黄视频在线播放免费| 久久久国产精品麻豆| 午夜日韩欧美国产| 国产一区二区三区在线臀色熟女| 国产成人精品久久二区二区免费| 中文字幕熟女人妻在线| 亚洲一区二区三区不卡视频| 久久精品国产亚洲av高清一级| www日本在线高清视频| 69av精品久久久久久| 成年版毛片免费区| 亚洲精华国产精华精| 亚洲 欧美 日韩 在线 免费| 亚洲成av人片免费观看| 老司机靠b影院| 中文字幕精品亚洲无线码一区|