Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 99
Filter
1.
Commun Biol ; 7(1): 1297, 2024 Oct 10.
Article in English | MEDLINE | ID: mdl-39390072

ABSTRACT

Colorectal cancer (CRC) ranks as the second most lethal cancer worldwide because of its high rate of metastasis, and approximately 20% of CRC patients have metastases at initial diagnosis. Metabolic reprogramming, a hallmark of cancer cells, has been implicated in the process of metastasis. We previously demonstrated that fucosyltransferase 2 (FUT2) promotes the malignancy of CRC cells, however, the underlying mechanisms remain unclear. Here, bioinformatic analysis revealed that FUT2 is associated with the malignant phenotype and fatty acid metabolism in CRC. FUT2 knockdown decreased glucose uptake and de novo fatty acid synthesis, which in turn inhibited the proliferation and metastasis of CRC cells. Mechanistically, FUT2 promotes YAP1 nuclear translocation and stabilizes mSREBP-1 by fucosylation, thus promoting de novo fatty acid synthesis in CRC cells. In summary, this study demonstrates that FUT2 promotes the proliferation and metastasis of CRC cells by reprogramming fatty acid metabolism via YAP/TAZ signaling and SREBP-1, indicating that FUT2 might be a potential target for developing therapeutic strategies against CRC.


Subject(s)
Colorectal Neoplasms , Fatty Acids , Fucosyltransferases , Galactoside 2-alpha-L-fucosyltransferase , Signal Transduction , Sterol Regulatory Element Binding Protein 1 , YAP-Signaling Proteins , Fucosyltransferases/genetics , Fucosyltransferases/metabolism , Colorectal Neoplasms/pathology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Humans , Fatty Acids/metabolism , Animals , Sterol Regulatory Element Binding Protein 1/metabolism , Sterol Regulatory Element Binding Protein 1/genetics , YAP-Signaling Proteins/metabolism , YAP-Signaling Proteins/genetics , Mice , Transcription Factors/metabolism , Transcription Factors/genetics , Cell Line, Tumor , Cell Proliferation , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Mice, Nude , Neoplasm Metastasis , Gene Expression Regulation, Neoplastic , Male , Female , Mice, Inbred BALB C , Transcriptional Coactivator with PDZ-Binding Motif Proteins/metabolism
2.
Anticancer Res ; 44(10): 4147-4153, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39348982

ABSTRACT

Epithelioid hemangioendothelioma (EHE) is a rare malignant vascular tumor arising from vascular endothelial cells. This study delves into the molecular mechanisms underlying EHE, with a specific focus on the Hippo-YAP/TAZ pathway. EHE is characterized molecularly by transcriptional co-activator with a PDZ-motif (TAZ)-calmodulin binding transcription activator 1 (CAMTA1) or Yes-associated protein (YAP)-transcription factor E3 (TFE3) fusions. YAP/TAZ, a transcription co-activator, binds to transcription factors and regulates gene expression. The YAP/TAZ and its upstream Hippo pathway are involved in cell proliferation and cell contact inhibition, regulating organ size and carcinogenesis. In addition to oncogenic effects, dysfunction or gene duplication of the Hippo pathway results in a poor prognosis due to epithelial-mesenchymal transformation of epithelial cells, stem cell transformation, and increased drug resistance. Notably, the TAZ-CAMTA1 fusion is specific to EHE, and genetic alterations in the Hippo pathway other than this fusion gene are absent in EHE. The TAZ-CAMTA1 fusion is a promising therapeutic target. This review summarizes recent advances in EHE, focusing on the role of the Hippo-YAP/TAZ pathway in EHE and its potential as a therapeutic target for drug development.


Subject(s)
Hemangioendothelioma, Epithelioid , Hippo Signaling Pathway , Protein Serine-Threonine Kinases , Signal Transduction , Transcription Factors , Transcriptional Coactivator with PDZ-Binding Motif Proteins , Humans , Hemangioendothelioma, Epithelioid/metabolism , Hemangioendothelioma, Epithelioid/pathology , Hemangioendothelioma, Epithelioid/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Transcriptional Coactivator with PDZ-Binding Motif Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Trans-Activators/metabolism , Trans-Activators/genetics , YAP-Signaling Proteins/metabolism , Molecular Targeted Therapy , Animals
3.
Sci Rep ; 14(1): 22233, 2024 09 27.
Article in English | MEDLINE | ID: mdl-39333200

ABSTRACT

Esophageal fibrosis can develop due to caustic or radiation injuries. Umbilical cord-derived mesenchymal stem cells (UC-MSCs) are known to mitigate fibrosis in various organs. However, the potential effects of UC-MSCs on human esophageal fibrosis remain underexplored. This study investigated the anti-fibrogenic properties and mechanisms of UC-MSC-derived conditioned media (UC-MSC-CM) on human esophageal fibroblasts (HEFs). HEFs were treated with TGF-ß1 and then cultured with UC-MSC-CM, and the expression levels of extracellular matrix (ECM) components, RhoA, myocardin related transcription factor A (MRTF-A), serum response factor (SRF), Yes-associated protein (YAP), and transcriptional coactivator with PDZ-binding motif (TAZ) were measured. UC-MSC-CM suppressed TGF-ß1-induced fibrogenic activation in HEFs, as evidenced by the downregulation of ECM. UC-MSC-CM diminished the expression of RhoA, MRTF-A, and SRF triggered by TGF-ß1. In TGF-ß1-stimulated HEFs, UC-MSC-CM decreased the nuclear localization of MRTF-A and YAP. Additionally, UC-MSC-CM diminished the TGF-ß1-induced nuclear expressions of YAP and TAZ, while concurrently enhancing the cytoplasmic presence of phosphorylated YAP. Furthermore, UC-MSC-CM reduced TGF-ß1-induced phosphorylation of Smad2. These findings suggest that UC-MSC-CM may inhibit TGF-ß1-induced fibrogenic activation in HEFs by targeting the Rho-mediated MRTF/SRF and YAP/TAZ pathways, as well as the Smad2 pathway. This indicates its potential as a stem cell therapy for esophageal fibrosis.


Subject(s)
Esophagus , Fibroblasts , Fibrosis , Mesenchymal Stem Cells , Trans-Activators , Transcription Factors , Transforming Growth Factor beta1 , rhoA GTP-Binding Protein , Humans , Mesenchymal Stem Cells/metabolism , Culture Media, Conditioned/pharmacology , Transforming Growth Factor beta1/metabolism , rhoA GTP-Binding Protein/metabolism , Esophagus/metabolism , Esophagus/cytology , Fibroblasts/metabolism , Trans-Activators/metabolism , Trans-Activators/genetics , Transcription Factors/metabolism , Umbilical Cord/cytology , YAP-Signaling Proteins/metabolism , Serum Response Factor/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Signal Transduction , Transcriptional Coactivator with PDZ-Binding Motif Proteins/metabolism , Cells, Cultured , Extracellular Matrix/metabolism , Smad2 Protein/metabolism
4.
Int J Mol Sci ; 25(18)2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39337493

ABSTRACT

Hippo-YAP/TAZ and Wnt/ß-catenin signaling pathways, by controlling proliferation, migration, cell fate, stemness, and apoptosis, are crucial regulators of development and tissue homeostasis. We employed zebrafish embryos as a model system to elucidate in living reporter organisms the crosstalk between the two signaling pathways. Co-expression analysis between the Wnt/ß-catenin Tg(7xTCF-Xla.Siam:GFP)ia4 and the Hippo-Yap/Taz Tg(Hsa.CTGF:nlsmCherry)ia49 zebrafish reporter lines revealed shared spatiotemporal expression profiles. These patterns were particularly evident in key developmental regions such as the midbrain-hindbrain boundary (MHB), epidermis, muscles, neural tube, notochord, floorplate, and otic vesicle. To investigate the relationship between the Wnt/ß-catenin pathway and Hippo-Yap/Taz signaling in vivo, we conducted a series of experiments employing both pharmacological and genetic strategies. Modulation of the Wnt/ß-catenin pathway with IWR-1, XAV939, or BIO resulted in a significant regulation of the Yap/Taz reporter signal, highlighting a clear correlation between ß-catenin and Yap/Taz activities. Furthermore, genetic perturbation of the Wnt/ß-catenin pathway, by APC inhibition or DKK1 upregulation, elicited evident and robust alteration of Yap/Taz activity. These findings revealed the intricate regulatory mechanisms underlying the crosstalk between the Wnt/ß-catenin and Hippo-Yap/Taz signaling, shedding light on their roles in orchestrating developmental processes in vivo.


Subject(s)
Embryonic Development , Gene Expression Regulation, Developmental , Wnt Signaling Pathway , YAP-Signaling Proteins , Zebrafish Proteins , Zebrafish , Animals , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish/embryology , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Embryonic Development/genetics , YAP-Signaling Proteins/metabolism , beta Catenin/metabolism , beta Catenin/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Transcriptional Coactivator with PDZ-Binding Motif Proteins/metabolism
5.
J Cell Sci ; 137(17)2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39140137

ABSTRACT

Mechanotransduction, which is the integration of mechanical signals from the external environment of a cell to changes in intracellular signaling, governs many cellular functions. Recent studies have shown that the mechanical state of the cell is also coupled to the cellular circadian clock. To investigate possible interactions between circadian rhythms and cellular mechanotransduction, we have developed a computational model that integrates the two pathways. We postulated that translocation of the transcriptional regulators MRTF (herein referring to both MRTF-A and MRTF-B), YAP and TAZ (also known as YAP1 and WWTR1, respectively; collectively denoted YAP/TAZ) into the nucleus leads to altered expression of circadian proteins. Simulations from our model predict that lower levels of cytoskeletal activity are associated with longer circadian oscillation periods and higher oscillation amplitudes, which is consistent with recent experimental observations. Furthermore, accumulation of YAP/TAZ and MRTF in the nucleus causes circadian oscillations to decay in our model. These effects hold both at the single-cell level and within a population-level framework. Finally, we investigated the effects of mutations in YAP or lamin A, the latter of which result in a class of diseases known as laminopathies. In silico, oscillations in circadian proteins are substantially weaker in populations of cells with mutations in YAP or lamin A, suggesting that defects in mechanotransduction can disrupt the circadian clock in certain disease states; however, reducing substrate stiffness in the model restores normal oscillatory behavior, suggesting a possible compensatory mechanism. Thus, our study identifies that mechanotransduction could be a potent modulatory cue for cellular clocks and that this crosstalk can be leveraged to rescue the circadian clock in disease states.


Subject(s)
Circadian Clocks , Mechanotransduction, Cellular , YAP-Signaling Proteins , Humans , Animals , YAP-Signaling Proteins/metabolism , Computer Simulation , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Trans-Activators/metabolism , Trans-Activators/genetics , Models, Biological , Cell Nucleus/metabolism , Mammals/metabolism , Transcriptional Coactivator with PDZ-Binding Motif Proteins/metabolism
6.
Matrix Biol ; 133: 103-115, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39153517

ABSTRACT

Skeletal muscle fibrosis is defined as the excessive accumulation of extracellular matrix (ECM) components and is a hallmark of muscular dystrophies. Fibro-adipogenic progenitors (FAPs) are the main source of ECM, and thus have been strongly implicated in fibrogenesis. In skeletal muscle fibrotic models, including muscular dystrophies, FAPs undergo dysregulations in terms of proliferation, differentiation, and apoptosis, however few studies have explored the impact of FAPs migration. Here, we studied fibroblast and FAPs migration and identified lysophosphatidic acid (LPA), a signaling lipid central to skeletal muscle fibrogenesis, as a significant migration inductor. We identified LPA receptor 1 (LPA1) mediated signaling as crucial for this effect through a mechanism dependent on the Hippo pathway, another pathway implicated in fibrosis across diverse tissues. This cross-talk favors the activation of the Yes-associated protein 1 (YAP) and Transcriptional coactivator with PDZ-binding motif (TAZ), leading to increased expression of fibrosis-associated genes. This study reveals the role of YAP in LPA-mediated fibrotic responses as inhibition of YAP transcriptional coactivator activity hinders LPA-induced migration in fibroblasts and FAPs. Moreover, we found that FAPs derived from the mdx4cv mice, a murine model of Duchenne muscular dystrophy, display a heightened migratory phenotype due to enhanced LPA signaling compared to wild-type FAPs. Remarkably, we found that the inhibition of LPA1 or YAP transcriptional coactivator activity in mdx4cv FAPs reverts this phenotype. In summary, the identified LPA-LPA1-YAP pathway emerges as a critical driver of skeletal muscle FAPs migration and provides insights into potential novel targets to mitigate fibrosis in muscular dystrophies.


Subject(s)
Adaptor Proteins, Signal Transducing , Cell Movement , Fibroblasts , Fibrosis , Lysophospholipids , Muscle, Skeletal , Receptors, Lysophosphatidic Acid , Signal Transduction , YAP-Signaling Proteins , Lysophospholipids/metabolism , Animals , YAP-Signaling Proteins/metabolism , YAP-Signaling Proteins/genetics , Mice , Receptors, Lysophosphatidic Acid/metabolism , Receptors, Lysophosphatidic Acid/genetics , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Fibroblasts/metabolism , Fibroblasts/pathology , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Humans , Hippo Signaling Pathway , Mice, Inbred mdx , Transcriptional Coactivator with PDZ-Binding Motif Proteins/metabolism , Adipogenesis/genetics , Muscular Dystrophies/metabolism , Muscular Dystrophies/genetics , Muscular Dystrophies/pathology
7.
Cancer Sci ; 115(10): 3370-3383, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39155534

ABSTRACT

The activation of yes-associated protein 1 (YAP1) and transcriptional co-activator with PDZ-binding motif (TAZ) has been implicated in both regeneration and tumorigenesis, thus representing a double-edged sword in tissue homeostasis. However, how the activity of YAP1/TAZ is regulated or what leads to its dysregulation in these processes remains unknown. To explore the upstream stimuli modulating the cellular activity of YAP1/TAZ, we developed a highly sensitive YAP1/TAZ/TEAD-responsive DNA element (YRE) and incorporated it into a lentivirus-based reporter cell system to allow for sensitive and specific monitoring of the endogenous activity of YAP1/TAZ in terms of luciferase activity in vitro and Venus fluorescence in vivo. Furthermore, by replacing YRE with TCF- and NF-κB-binding DNA elements, we demonstrated the applicability of this reporter system to other pathways such as Wnt/ß-catenin/TCF- and IL-1ß/NF-κB-mediated signaling, respectively. The practicality of this system was evaluated by performing cell-based reporter screening of a chemical compound library consisting of 364 known inhibitors, using reporter-introduced cells capable of quantifying YAP1/TAZ- and ß-catenin-mediated transcription activities, which led to the identification of multiple inhibitors, including previously known as well as novel modulators of these signaling pathways. We further confirmed that novel YAP1/TAZ modulators, such as potassium ionophores, Janus kinase inhibitors, platelet-derived growth factor receptor inhibitors, and genotoxic stress inducers, alter the protein level or phosphorylation of endogenous YAP1/TAZ and the expression of their target genes. Thus, this reporter system provides a powerful tool to monitor endogenous signaling activities of interest (even in living cells) and search for modulators in various cellular contexts.


Subject(s)
Adaptor Proteins, Signal Transducing , Genes, Reporter , Phosphoproteins , Transcription Factors , YAP-Signaling Proteins , Humans , Adaptor Proteins, Signal Transducing/metabolism , Transcription Factors/metabolism , YAP-Signaling Proteins/metabolism , Phosphoproteins/metabolism , Transcriptional Coactivator with PDZ-Binding Motif Proteins/metabolism , Trans-Activators/metabolism , NF-kappa B/metabolism , Signal Transduction , beta Catenin/metabolism , HEK293 Cells , Response Elements , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics
8.
ACS Biomater Sci Eng ; 10(9): 5550-5561, 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39190867

ABSTRACT

The transcriptional coactivators yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) are master regulators involved in a multitude of cancer types and a wide range of tumorigenic events, including cancer stem cell renewal, invasion, metastasis, tumor precursor emergence, and drug resistance. YAP/TAZ are known to be regulated by several external cues and stimuli, such as extracellular matrix stiffness, cell spreading, cell geometry, and shear stress. Therefore, there is a need in the field of cancer research to develop and design relevant in vitro models that can accurately reflect the complex biochemical and biophysical cues of the tumor microenvironment central to the YAP/TAZ signaling nexus. While much progress has been made, this remains a major roadblock to advancing research in this field. In this review, we highlight the current engineered biomaterials and in vitro model systems that can be used to advance our understanding of how YAP/TAZ shapes several aspects of cancer. We begin by discussing current 2D and 3D hydrogel systems that model the YAP/TAZ response to ECM stiffness. We then examine the current trends in organoid culture systems and the use of microfluidics to model the effects of cellular density and shear stress on YAP/TAZ. Finally, we analyze the ongoing pitfalls of the present models used and important future directions in engineering systems that will advance our current knowledge of YAP/TAZ in cancer.


Subject(s)
Biocompatible Materials , Neoplasms , Transcription Factors , Humans , Neoplasms/metabolism , Neoplasms/pathology , Transcription Factors/metabolism , YAP-Signaling Proteins/metabolism , Transcriptional Coactivator with PDZ-Binding Motif Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Hydrogels/chemistry , Tumor Microenvironment , Animals , Trans-Activators/metabolism , Models, Biological
9.
Nat Commun ; 15(1): 5809, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38987584

ABSTRACT

Human papillomaviruses (HPVs) cause most cervical cancers and an increasing number of anogenital and oral carcinomas, with most cases caused by HPV16 or HPV18. HPV hijacks host signalling pathways to promote carcinogenesis. Understanding these interactions could permit identification of much-needed therapeutics for HPV-driven malignancies. The Hippo signalling pathway is important in HPV+ cancers, with the downstream effector YAP playing a pro-oncogenic role. In contrast, the significance of its paralogue TAZ remains largely uncharacterised in these cancers. We demonstrate that TAZ is dysregulated in a HPV-type dependent manner by a distinct mechanism to that of YAP and controls proliferation via alternative cellular targets. Analysis of cervical cancer cell lines and patient biopsies revealed that TAZ expression was only significantly increased in HPV18+ and HPV18-like cells and TAZ knockdown reduced proliferation, migration and invasion only in HPV18+ cells. RNA-sequencing of HPV18+ cervical cells revealed that YAP and TAZ have distinct targets, suggesting they promote carcinogenesis by different mechanisms. Thus, in HPV18+ cancers, YAP and TAZ play non-redundant roles. This analysis identified TOGARAM2 as a previously uncharacterised TAZ target and demonstrates its role as a key effector of TAZ-mediated proliferation, migration and invasion in HPV18+ cancers.


Subject(s)
Adaptor Proteins, Signal Transducing , Cell Proliferation , Hippo Signaling Pathway , Human papillomavirus 18 , Papillomavirus Infections , Protein Serine-Threonine Kinases , Signal Transduction , Transcription Factors , Uterine Cervical Neoplasms , YAP-Signaling Proteins , Female , Humans , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Carcinogenesis/genetics , Cell Line, Tumor , Cell Movement , Gene Expression Regulation, Neoplastic , Human papillomavirus 16/genetics , Human papillomavirus 16/metabolism , Human papillomavirus 18/genetics , Human papillomavirus 18/metabolism , Papillomavirus Infections/virology , Papillomavirus Infections/metabolism , Papillomavirus Infections/genetics , Papillomavirus Infections/pathology , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Trans-Activators/metabolism , Trans-Activators/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Transcriptional Coactivator with PDZ-Binding Motif Proteins/metabolism , Uterine Cervical Neoplasms/virology , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/pathology , YAP-Signaling Proteins/metabolism
10.
Adv Sci (Weinh) ; 11(34): e2401588, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38981023

ABSTRACT

Colorectal cancer (CRC) and inflammatory bowel disease (IBD) are escalating global health concerns. Despite their distinct clinical presentations, both disorders share intricate genetic and molecular interactions. The Hippo signaling pathway plays a crucial role in regulating cell processes and is implicated in the pathogenesis of IBD and CRC. Circular RNAs (circRNAs) have gained attention for their roles in various diseases, including IBD and CRC. However, a comprehensive understanding of specific circRNAs involved in both IBD and CRC, and their functional roles is lacking. Here, it is found that circHIPK2 (hsa_circRNA_104507) is a bona fide circRNA consistently upregulated in both IBD and CRC suggesting its potential as a biomarker. Furthermore, silencing of circHIPK2 suppressed the growth of CRC cells in vitro and in vivo. Interestingly, decreased circHipk2 potentiated dextran sulfate sodium (DSS)-induced colitis but alleviated colitis-associated tumorigenesis. Most significantly, mechanistic investigations further unveil that circHIPK2, mediated by FUS, interacting with EIF4A3 to promote the translation of TAZ, ultimately increasing the transcription of downstream target genes CCN1 and CCN2. Taken together, circHIPK2 emerges as a key player in the shared mechanisms of IBD and CRC, modulating the Hippo signaling pathway. CircHIPK2-EIF4A3 axis contributes to cell growth in intestinal epithelial of colitis and CRC by enhancing TAZ translation.


Subject(s)
Colitis , Colorectal Neoplasms , Protein Serine-Threonine Kinases , RNA, Circular , Transcriptional Coactivator with PDZ-Binding Motif Proteins , RNA, Circular/genetics , RNA, Circular/metabolism , Colitis/genetics , Colitis/metabolism , Colitis/chemically induced , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Humans , Mice , Animals , Transcriptional Coactivator with PDZ-Binding Motif Proteins/metabolism , Transcriptional Coactivator with PDZ-Binding Motif Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Cell Proliferation/genetics , Disease Models, Animal , Intestinal Mucosa/metabolism
11.
Exp Hematol ; 138: 104282, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39032857

ABSTRACT

We and others have previously shown that TAZ plays a tumor suppressive role in multiple myeloma. However, recent reports suggest that molecular crosstalk between the myeloma cells and bone marrow stromal components contributes to the myeloma cell survival and drug resistance. These reports further point to reciprocal interaction via adhesion molecules as the most prominent mechanism of intercellular crosstalk between myeloma cells and bone marrow mesenchymal stromal cells (BM-MSCs). YAP/TAZ silencing/expression has been shown to correlate across all cancers with a set of adhesion/extracellular matrix proteins. Therefore, we hypothesized that TAZ may regulate myeloma cell interaction with BM stromal cells by influencing the expression of distinct cell adhesion signatures. We used previously established TAZ myeloma cell line models, including DELTA47-pLENTI or TAZ knockout DELTA47 cells cocultured with or without BM-MSCs, as our study models. Using RNA sequencing analysis, we performed the first comprehensive screen for cell adhesion-related transcriptional targets of TAZ in multiple myeloma (MM). In doing so, we uncovered an enrichment of cell adhesion-related genes in TAZ knockout DELTA47 cells relatively to pLENTI-DELTA47 cells, including 11 genes with log2 fold change > 2 (p < 0.05), namely, ANXA1, ADGRL2, NCAM1, NCAM2, ADGRL3, CXADR, ALCAM, JAM2, KIRREL1, KIRREL2, and ADGRG7, suggesting possible relationship with TAZ. We validated ANXA1 as a bona fide target of TAZ in MM. We show that TAZ represses myeloma cell migration and interaction with BM-MSCs by transcriptionally downregulating ANXA1 expression via TEAD-dependent mechanism. Our data provide new insights into the understanding of the role of TAZ in the intercellular communication signals between myeloma cells and BM-MSCs. Our findings also suggest that ANXA1 represents a putative cell adhesion target to attenuate BM-MSC driven, tumor-promoting interaction with myeloma cells.


Subject(s)
Annexin A1 , Cell Communication , Down-Regulation , Gene Expression Regulation, Neoplastic , Mesenchymal Stem Cells , Multiple Myeloma , Transcriptional Coactivator with PDZ-Binding Motif Proteins , Multiple Myeloma/pathology , Multiple Myeloma/metabolism , Multiple Myeloma/genetics , Humans , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Transcriptional Coactivator with PDZ-Binding Motif Proteins/metabolism , Cell Line, Tumor , Annexin A1/genetics , Annexin A1/metabolism , Cell Adhesion , Coculture Techniques , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasm Proteins/biosynthesis
12.
Elife ; 132024 Jul 25.
Article in English | MEDLINE | ID: mdl-39051998

ABSTRACT

The Hippo pathway plays a central role in tissue development and homeostasis. However, the function of Hippo in pancreatic endocrine development remains obscure. Here, we generated novel conditional genetically engineered mouse models to examine the roles of Hippo pathway-mediated YAP1/TAZ inhibition in the development stages of endocrine specification and differentiation. While YAP1 protein was localized to the nuclei in bipotent progenitor cells, Neurogenin 3 expressing endocrine progenitors completely lost YAP1 expression. Using genetically engineered mouse models, we found that inactivation of YAP1 requires both an intact Hippo pathway and Neurogenin 3 protein. Gene deletion of Lats1 and 2 kinases (Lats1&2) in endocrine progenitor cells of developing mouse pancreas using Neurog3Cre blocked endocrine progenitor cell differentiation and specification, resulting in reduced islets size and a disorganized pancreas at birth. Loss of Lats1&2 in Neurogenin 3 expressing cells activated YAP1/TAZ transcriptional activity and recruited macrophages to the developing pancreas. These defects were rescued by deletion of Yap1/Wwtr1 genes, suggesting that tight regulation of YAP1/TAZ by Hippo signaling is crucial for pancreatic endocrine specification. In contrast, deletion of Lats1&2 using ß-cell-specific Ins1CreER resulted in a phenotypically normal pancreas, indicating that Lats1&2 are indispensable for differentiation of endocrine progenitors but not for that of ß-cells. Our results demonstrate that loss of YAP1/TAZ expression in the pancreatic endocrine compartment is not a passive consequence of endocrine specification. Rather, Hippo pathway-mediated inhibition of YAP1/TAZ in endocrine progenitors is a prerequisite for endocrine specification and differentiation.


Subject(s)
Adaptor Proteins, Signal Transducing , Cell Differentiation , Protein Serine-Threonine Kinases , Signal Transduction , YAP-Signaling Proteins , Animals , YAP-Signaling Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Mice , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Hippo Signaling Pathway , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Trans-Activators/metabolism , Trans-Activators/genetics , Islets of Langerhans/metabolism , Islets of Langerhans/embryology , Transcription Factors/metabolism , Transcription Factors/genetics , Transcriptional Coactivator with PDZ-Binding Motif Proteins/metabolism , Phosphoproteins/metabolism , Phosphoproteins/genetics , Acyltransferases , Tumor Suppressor Proteins
13.
Elife ; 122024 Jul 24.
Article in English | MEDLINE | ID: mdl-39046443

ABSTRACT

The role of processing bodies (P-bodies) in tumorigenesis and tumor progression is not well understood. Here, we showed that the oncogenes YAP/TAZ promote P-body formation in a series of cancer cell lines. Mechanistically, both transcriptional activation of the P-body-related genes SAMD4A, AJUBA, and WTIP and transcriptional suppression of the tumor suppressor gene PNRC1 are involved in enhancing the effects of YAP/TAZ on P-body formation in colorectal cancer (CRC) cells. By reexpression of PNRC1 or knockdown of P-body core genes (DDX6, DCP1A, and LSM14A), we determined that disruption of P-bodies attenuates cell proliferation, cell migration, and tumor growth induced by overexpression of YAP5SA in CRC. Analysis of a pancancer CRISPR screen database (DepMap) revealed co-dependencies between YAP/TEAD and the P-body core genes and correlations between the mRNA levels of SAMD4A, AJUBA, WTIP, PNRC1, and YAP target genes. Our study suggests that the P-body is a new downstream effector of YAP/TAZ, which implies that reexpression of PNRC1 or disruption of P-bodies is a potential therapeutic strategy for tumors with active YAP.


Subject(s)
Adaptor Proteins, Signal Transducing , Carcinogenesis , Trans-Activators , Transcription Factors , Transcriptional Coactivator with PDZ-Binding Motif Proteins , YAP-Signaling Proteins , Humans , YAP-Signaling Proteins/metabolism , YAP-Signaling Proteins/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Carcinogenesis/genetics , Cell Line, Tumor , Transcriptional Coactivator with PDZ-Binding Motif Proteins/metabolism , Trans-Activators/metabolism , Trans-Activators/genetics , Animals , Cell Proliferation , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Mice , Phosphoproteins/metabolism , Phosphoproteins/genetics , Gene Expression Regulation, Neoplastic , Cell Movement , LIM Domain Proteins
14.
Arch Pharm Res ; 47(6): 558-570, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38874747

ABSTRACT

Metabolic dysfunction-associated steatotic liver disease (MASLD) is becoming an increasingly pressing global health challenge, with increasing mortality rates showing an upward trend. Two million deaths occur annually from cirrhosis and liver cancer together each year. Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ), key effectors of the Hippo signaling pathway, critically regulate tissue homeostasis and disease progression in the liver. While initial studies have shown that YAP expression is normally restricted to cholangiocytes in healthy livers, the activation of YAP/TAZ is observed in other hepatic cells during chronic liver disease. The disease-driven dysregulation of YAP/TAZ appears to be a critical element in the MASLD progression, contributing to hepatocyte dysfunction, inflammation, and fibrosis. In this study, we focused on the complex roles of YAP/TAZ in MASLD and explored how the YAP/TAZ dysregulation of YAP/TAZ drives steatosis, inflammation, fibrosis, and cirrhosis. Finally, the cell-type-specific functions of YAP/TAZ in different types of hepatic cells, such as hepatocytes, hepatic stellate cells, hepatic macrophages, and biliary epithelial cells are discussed, highlighting the multifaceted impact of YAP/TAZ on liver physiology and pathology.


Subject(s)
Adaptor Proteins, Signal Transducing , Disease Progression , Transcription Factors , Transcriptional Coactivator with PDZ-Binding Motif Proteins , YAP-Signaling Proteins , Humans , Transcription Factors/metabolism , YAP-Signaling Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Transcriptional Coactivator with PDZ-Binding Motif Proteins/metabolism , Fatty Liver/metabolism , Fatty Liver/pathology , Trans-Activators/metabolism , Signal Transduction
15.
Int J Oncol ; 65(2)2024 Aug.
Article in English | MEDLINE | ID: mdl-38873993

ABSTRACT

Genes encoding subunits of SWI/SNF (BAF) chromatin­remodeling complexes are recurrently mutated in a broad array of tumor types, and among the subunits, ARID1A is the most frequent target with mutations. In the present study, it was reported that ARID1A inhibits the epithelial­mesenchymal transition (EMT) and stemness of ovarian cancer cells, accompanied by reduced cell viability, migration and colony formation, suggesting that ARID1A acts as a tumor suppressor in ovarian cancer. Mechanistically, ARID1A exerts its inhibitory effects on ovarian cancer cells by activating the Hippo signaling pathway. Conversely, the overexpression of a gain­of­function transcriptional co­activator with PDZ­binding motif (TAZ) mutant (TAZ­Ser89) effectively reverses the effects induced by ARID1A. In addition, activation of Hippo signaling apparently upregulates ARID1A protein expression, whereas ectopic expression of TAZ­Ser89 results in the markedly decreased ARID1A levels, indicating a feedback of ARID1A­TAZ in regulating ovarian cancer cell EMT and stemness. Thus, the present study uncovered the role of ARID1A through the Hippo/TAZ pathway in modulating EMT and stemness of ovarian cancer cells, and providing with evidence that TAZ inhibitors could effectively prevent initiation and metastasis of ovarian cancer cases where ARID1A is lost or mutated.


Subject(s)
Epithelial-Mesenchymal Transition , Hippo Signaling Pathway , Neoplastic Stem Cells , Ovarian Neoplasms , Transcription Factors , Female , Humans , Cell Line, Tumor , Cell Movement , Cell Proliferation , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Signal Transduction , Transcription Factors/metabolism , Transcription Factors/genetics , Transcriptional Coactivator with PDZ-Binding Motif Proteins/metabolism
16.
Mol Aspects Med ; 98: 101280, 2024 08.
Article in English | MEDLINE | ID: mdl-38870717

ABSTRACT

Although cancer diagnosis and treatment have rapidly advanced in recent decades, urological malignancies, which have high morbidity and mortality rates, are among the most difficult diseases to treat. The Hippo signaling is an evolutionarily conserved pathway in organ size control and tissue homeostasis maintenance. Its downstream effectors, Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ), are key modulators of numerous physiological and pathological processes. Recent work clearly indicates that Hippo signaling is frequently altered in human urological malignancies. In this review, we discuss the disparate viewpoints on the upstream regulators of YAP/TAZ and their downstream targets and systematically summarize the biological implications. More importantly, we highlight the molecular mechanisms involved in Hippo-YAP signaling to improve our understanding of its role in every stage of prostate cancer, bladder cancer and kidney cancer progression. A better understanding of the biological outcomes of YAP/TAZ modulation will contribute to the establishment of future therapeutic approaches.


Subject(s)
Hippo Signaling Pathway , Protein Serine-Threonine Kinases , Signal Transduction , Transcription Factors , Humans , Protein Serine-Threonine Kinases/metabolism , Transcription Factors/metabolism , Urologic Neoplasms/metabolism , Urologic Neoplasms/etiology , Urologic Neoplasms/pathology , YAP-Signaling Proteins/metabolism , Animals , Transcriptional Coactivator with PDZ-Binding Motif Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Gene Expression Regulation, Neoplastic , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/pathology , Urinary Bladder Neoplasms/genetics , Male
17.
FASEB J ; 38(10): e23636, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38752683

ABSTRACT

Osteoarthritis (OA) and rheumatoid arthritis (RA) are two common forms of arthritis with undefined etiology and pathogenesis. Yes-associated protein (YAP) and its homolog transcriptional coactivator with PDZ-binding motif (TAZ), which act as sensors for cellular mechanical and inflammatory cues, have been identified as crucial players in the regulation of joint homeostasis. Current studies also reveal a significant association between YAP/TAZ and the pathogenesis of OA and RA. The objective of this review is to elucidate the impact of YAP/TAZ on different joint tissues and to provide inspiration for further studying the potential therapeutic implications of YAP/TAZ on arthritis. Databases, such as PubMed, Cochran Library, and Embase, were searched for all available studies during the past two decades, with keywords "YAP," "TAZ," "OA," and "RA."


Subject(s)
Adaptor Proteins, Signal Transducing , Arthritis, Rheumatoid , Osteoarthritis , Transcription Factors , YAP-Signaling Proteins , Humans , Transcription Factors/metabolism , Animals , Arthritis, Rheumatoid/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , YAP-Signaling Proteins/metabolism , Osteoarthritis/metabolism , Osteoarthritis/etiology , Transcriptional Coactivator with PDZ-Binding Motif Proteins/metabolism , Joints/metabolism , Joints/pathology , Trans-Activators/metabolism , Trans-Activators/genetics
18.
Respir Res ; 25(1): 212, 2024 May 18.
Article in English | MEDLINE | ID: mdl-38762455

ABSTRACT

Paraquat (PQ) is a widely used herbicide and a common cause of poisoning that leads to pulmonary fibrosis with a high mortality rate. However, the underlying mechanisms of PQ-induced pulmonary fibrosis and whether pulmonary epithelial cell senescence is involved in the process remain elusive. In this study, PQ-induced pulmonary epithelial cell senescence and Hippo-YAP/TAZ activation were observed in both C57BL/6 mice and human epithelial cells. PQ-induced senescent pulmonary epithelial cells promoted lung fibroblast transformation through secreting senescence-associated secretory phenotype (SASP) factors. Yap/Taz knockdown in mice lungs significantly decreased the expression of downstream profibrotic protein Ctgf and senescent markers p16 and p21, and alleviated PQ-induced pulmonary fibrosis. Interfering YAP/TAZ in senescent human pulmonary epithelial cells resulted in decreased expression of the anti-apoptosis protein survivin and elevated level of apoptosis. In conclusion, our findings reveal a novel mechanism by which the involvement of Hippo-YAP/TAZ activation in pulmonary epithelial cell senescence mediates the pathogenesis of PQ-induced pulmonary fibrosis, thereby offering novel insights and potential targets for the clinical management of PQ poisoning as well as providing the mechanistic insight of the involvement of Yap/Taz activation in cell senescence in pulmonary fibrosis and its related pulmonary disorders. The YIN YANG balance between cell senescence and apoptosis is important to maintain the homeostasis of the lung, the disruption of which will lead to disease.


Subject(s)
Adaptor Proteins, Signal Transducing , Cellular Senescence , Mice, Inbred C57BL , Paraquat , Pulmonary Fibrosis , Transcription Factors , YAP-Signaling Proteins , Animals , Cellular Senescence/drug effects , Cellular Senescence/physiology , YAP-Signaling Proteins/metabolism , Humans , Mice , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/pathology , Adaptor Proteins, Signal Transducing/metabolism , Transcription Factors/metabolism , Transcription Factors/genetics , Paraquat/toxicity , Male , Transcriptional Coactivator with PDZ-Binding Motif Proteins/metabolism , Epithelial Cells/metabolism , Epithelial Cells/drug effects , Epithelial Cells/pathology , Trans-Activators/metabolism , Trans-Activators/genetics
19.
Int J Biol Sci ; 20(7): 2592-2606, 2024.
Article in English | MEDLINE | ID: mdl-38725855

ABSTRACT

Transcriptional coactivator with a PDZ-binding motif (TAZ) plays a key role in normal tissue homeostasis and tumorigenesis through interaction with several transcription factors. In particular, TAZ deficiency causes abnormal alveolarization and emphysema, and persistent TAZ overexpression contributes to lung cancer and pulmonary fibrosis, suggesting the possibility of a complex mechanism of TAZ function. Recent studies suggest that nuclear factor erythroid 2-related factor 2 (NRF2), an antioxidant defense system, induces TAZ expression during tumorigenesis and that TAZ also activates the NRF2-mediated antioxidant pathway. We thus thought to elucidate the cross-regulation of TAZ and NRF2 and the underlying molecular mechanisms and functions. TAZ directly interacted with NRF2 through the N-terminal domain and suppressed the transcriptional activity of NRF2 by preventing NRF2 from binding to DNA. In addition, the return of NRF2 to basal levels after signaling was inhibited in TAZ deficiency, resulting in sustained nuclear NRF2 levels and aberrantly increased expression of NRF2 targets. TAZ deficiency failed to modulate optimal NRF2 signaling and concomitantly impaired lysosomal acidification and lysosomal enzyme function, accumulating the abnormal autophagy vesicles and reactive oxygen species and causing protein oxidation and cellular damage in the lungs. TAZ restoration to TAZ deficiency normalized dysregulated NRF2 signaling and aberrant lysosomal function and triggered the normal autophagy-lysosomal pathway. Therefore, TAZ is indispensable for the optimal regulation of NRF2-mediated autophagy-lysosomal pathways and for preventing pulmonary damage caused by oxidative stress and oxidized proteins.


Subject(s)
Autophagy , Lysosomes , NF-E2-Related Factor 2 , NF-E2-Related Factor 2/metabolism , Autophagy/physiology , Lysosomes/metabolism , Animals , Mice , Humans , Transcriptional Coactivator with PDZ-Binding Motif Proteins/metabolism , Signal Transduction , Transcription Factors/metabolism , Transcription Factors/genetics , Reactive Oxygen Species/metabolism , Adaptor Proteins, Signal Transducing
20.
Nat Metab ; 6(5): 847-860, 2024 May.
Article in English | MEDLINE | ID: mdl-38811804

ABSTRACT

Adipose tissues serve as an energy reservoir and endocrine organ, yet the mechanisms that coordinate these functions remain elusive. Here, we show that the transcriptional coregulators, YAP and TAZ, uncouple fat mass from leptin levels and regulate adipocyte plasticity to maintain metabolic homeostasis. Activating YAP/TAZ signalling in adipocytes by deletion of the upstream regulators Lats1 and Lats2 results in a profound reduction in fat mass by converting mature adipocytes into delipidated progenitor-like cells, but does not cause lipodystrophy-related metabolic dysfunction, due to a paradoxical increase in circulating leptin levels. Mechanistically, we demonstrate that YAP/TAZ-TEAD signalling upregulates leptin expression by directly binding to an upstream enhancer site of the leptin gene. We further show that YAP/TAZ activity is associated with, and functionally required for, leptin regulation during fasting and refeeding. These results suggest that adipocyte Hippo-YAP/TAZ signalling constitutes a nexus for coordinating adipose tissue lipid storage capacity and systemic energy balance through the regulation of adipocyte plasticity and leptin gene transcription.


Subject(s)
Adaptor Proteins, Signal Transducing , Adipocytes , Adipose Tissue , Energy Metabolism , Hippo Signaling Pathway , Leptin , Protein Serine-Threonine Kinases , Signal Transduction , YAP-Signaling Proteins , Animals , Leptin/metabolism , Protein Serine-Threonine Kinases/metabolism , Mice , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , YAP-Signaling Proteins/metabolism , Adipose Tissue/metabolism , Adipocytes/metabolism , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Transcriptional Coactivator with PDZ-Binding Motif Proteins/metabolism , Phosphoproteins/metabolism , Phosphoproteins/genetics , Tumor Suppressor Proteins/metabolism , Tumor Suppressor Proteins/genetics , Trans-Activators/metabolism , Trans-Activators/genetics
SELECTION OF CITATIONS
SEARCH DETAIL