1
|
Hashemolhosseini S, Gessler L. Crosstalk among canonical Wnt and Hippo pathway members in skeletal muscle and at the neuromuscular junction. Neural Regen Res 2025; 20:2464-2479. [PMID: 39248171 PMCID: PMC11801303 DOI: 10.4103/nrr.nrr-d-24-00417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 07/04/2024] [Accepted: 08/05/2024] [Indexed: 09/10/2024] Open
Abstract
Skeletal muscles are essential for locomotion, posture, and metabolic regulation. To understand physiological processes, exercise adaptation, and muscle-related disorders, it is critical to understand the molecular pathways that underlie skeletal muscle function. The process of muscle contraction, orchestrated by a complex interplay of molecular events, is at the core of skeletal muscle function. Muscle contraction is initiated by an action potential and neuromuscular transmission requiring a neuromuscular junction. Within muscle fibers, calcium ions play a critical role in mediating the interaction between actin and myosin filaments that generate force. Regulation of calcium release from the sarcoplasmic reticulum plays a key role in excitation-contraction coupling. The development and growth of skeletal muscle are regulated by a network of molecular pathways collectively known as myogenesis. Myogenic regulators coordinate the differentiation of myoblasts into mature muscle fibers. Signaling pathways regulate muscle protein synthesis and hypertrophy in response to mechanical stimuli and nutrient availability. Several muscle-related diseases, including congenital myasthenic disorders, sarcopenia, muscular dystrophies, and metabolic myopathies, are underpinned by dysregulated molecular pathways in skeletal muscle. Therapeutic interventions aimed at preserving muscle mass and function, enhancing regeneration, and improving metabolic health hold promise by targeting specific molecular pathways. Other molecular signaling pathways in skeletal muscle include the canonical Wnt signaling pathway, a critical regulator of myogenesis, muscle regeneration, and metabolic function, and the Hippo signaling pathway. In recent years, more details have been uncovered about the role of these two pathways during myogenesis and in developing and adult skeletal muscle fibers, and at the neuromuscular junction. In fact, research in the last few years now suggests that these two signaling pathways are interconnected and that they jointly control physiological and pathophysiological processes in muscle fibers. In this review, we will summarize and discuss the data on these two pathways, focusing on their concerted action next to their contribution to skeletal muscle biology. However, an in-depth discussion of the non-canonical Wnt pathway, the fibro/adipogenic precursors, or the mechanosensory aspects of these pathways is not the focus of this review.
Collapse
Affiliation(s)
- Said Hashemolhosseini
- Institute of Biochemistry, Medical Faculty, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | - Lea Gessler
- Institute of Biochemistry, Medical Faculty, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
2
|
Haripriya E, Hemalatha K, Matada GSP, Pal R, Das PK, Ashadul Sk MD, Mounika S, Viji MP, Aayishamma I, Jayashree KR. Advancements of anticancer agents by targeting the Hippo signalling pathway: biological activity, selectivity, docking analysis, and structure-activity relationship. Mol Divers 2025; 29:2829-2862. [PMID: 39436581 DOI: 10.1007/s11030-024-11009-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 09/30/2024] [Indexed: 10/23/2024]
Abstract
The Hippo signalling pathway is prominent and governs cell proliferation and stem cell activity, acting as a growth regulator and tumour suppressor. Defects in Hippo signalling and hyperactivation of its downstream effector's Yes-associated protein (YAP) and transcriptional co-activator with PDZ-binding motif (TAZ) play roles in cancer development, implying that pharmacological inhibition of YAP and TAZ activity could be an effective cancer treatment strategy. Conversely, YAP and TAZ can also have beneficial effects in promoting tissue repair and regeneration following damage, therefore their activation may be therapeutically effective in certain instances. Recently, a complex network of intracellular and extracellular signalling mechanisms that affect YAP and TAZ activity has been uncovered. The YAP/TAZ-TEAD interaction leads to tumour development and the protein structure of YAP/TAZ-TEAD includes three interfaces and one hydrophobic pocket. There are clinical and preclinical trial drugs available to inhibit the hippo signalling pathway, but these drugs have moderate to severe side effects, so researchers are in search of novel, potent, and selective hippo signalling pathway inhibitors. In this review, we have discussed the hippo pathway in detail, including its structure, activation, and role in cancer. We have also provided the various inhibitors under clinical and preclinical trials, and advancement of small molecules their detailed docking analysis, structure-activity relationship, and biological activity. We anticipate that the current study will be a helpful resource for researchers.
Collapse
Affiliation(s)
- E Haripriya
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru, 560107, Karnataka, India
| | - K Hemalatha
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru, 560107, Karnataka, India.
| | - Gurubasavaraja Swamy Purawarga Matada
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru, 560107, Karnataka, India
| | - Rohit Pal
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru, 560107, Karnataka, India.
| | - Pronoy Kanti Das
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru, 560107, Karnataka, India
| | - M D Ashadul Sk
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru, 560107, Karnataka, India
| | - S Mounika
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru, 560107, Karnataka, India
| | - M P Viji
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru, 560107, Karnataka, India
| | - I Aayishamma
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru, 560107, Karnataka, India
| | - K R Jayashree
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru, 560107, Karnataka, India
| |
Collapse
|
3
|
Uttagomol J, Wongviriya A, Chantaramanee A, Prasitsak T. YAP Expression is Related to the Aggressive Behavior of Odontogenic Cysts. Eur J Dent 2025. [PMID: 40311635 DOI: 10.1055/s-0044-1801275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2025] Open
Abstract
OBJECTIVE Yes-associated protein (YAP) and Ki-67 are known to be involved in cell proliferation. While their overexpression is associated with aggressive behaviors in oral squamous cell carcinoma (OSCC), their roles in odontogenic cysts have not been thoroughly investigated. Therefore, this study aimed to evaluate the immunohistochemical expression of YAP and Ki-67 in odontogenic cysts. MATERIAL AND METHODS Tissue samples included 12 radicular cysts, 10 dentigerous cysts, 9 odontogenic keratocysts, and 9 normal oral mucosa specimens. Immunohistochemical analysis was conducted both manually and using ImageJ software for comparison. STATISTICAL ANALYSIS The Kruskal-Wallis, followed by the Mann-Whitney U-test was used to assess the differences in the YAP and Ki-67 immunoexpression levels among various group lesions. The correlation between those two proteins in individual groups was tested by Spearman correlation. A p-value of less than 0.05 was considered statistically significant. SPSS software version 25.0 was used. RESULTS There was a strong nuclear staining of YAP in basal and superficial cells and an intense cytoplasmic YAP positivity in odontogenic keratocysts, whereas a weaker YAP staining in both the nucleus and cytoplasm throughout the epithelial thickness was observed in dentigerous cyst, radicular cyst, and normal oral mucosa, respectively. YAP expression differed significantly in all odontogenic cysts compared with normal oral mucosa (p < 0.05). Ki-67 expression was notably higher in odontogenic keratocysts relative to other cysts and normal oral mucosa. Although no statistically significant correlation was found between YAP and Ki-67 across the groups, both proteins displayed similar positive trends in odontogenic keratocysts. CONCLUSION These findings suggest that YAP activation may be related to the proliferative behavior of odontogenic cysts, especially in more aggressive lesions, but less likely to influence the inflammatory cysts. This insight could improve understanding of their pathogenesis and pave the way for new diagnostic and therapeutic approaches.
Collapse
Affiliation(s)
- Jutamas Uttagomol
- Department of Oral Diagnosis, Faculty of Dentistry, Naresuan University, Phitsanulok, Thailand
| | - Adjabhak Wongviriya
- Department of Oral Diagnosis, Faculty of Dentistry, Naresuan University, Phitsanulok, Thailand
| | - Ariya Chantaramanee
- Department of Preventive Dentistry, Faculty of Dentistry, Naresuan University, Phitsanulok, Thailand
| | - Thanit Prasitsak
- Department of Oral Biology, Faculty of Dentistry, Naresuan University, Phitsanulok, Thailand
| |
Collapse
|
4
|
İnanç İ, Bender O, Atalay A, Köse SK, Erdemli E. Alterations in the Hippo Signaling Pathway During Adenogenesis Impairment in Postnatal Mouse Uterus. Reprod Sci 2025; 32:1685-1698. [PMID: 40106220 PMCID: PMC12041100 DOI: 10.1007/s43032-025-01793-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 01/11/2025] [Indexed: 03/22/2025]
Abstract
The mouse uterus, which consists of single-layered epithelium and undifferentiated mesenchyme at birth, begins to differentiate in the postnatal period. The process of adenogenesis, defined as gland development, begins on the Postnatal (PN) Day 5, and this process is very evident on the PN Day 10. Although various signaling pathways effective in the adenogenesis process but the mechanism underlying this progress have not been clarified yet. Hippo signaling pathway have roles in many cellular functions, such as proliferation, differentiation and cell death. But the relationship between the Hippo signaling pathway and uterus adenogenesis is unknown. The objective of this study has been to determine if there is a change in the Hippo signaling pathway in mice with impaired gland development during the adenogenesis process. To that aim, we use mouse uterus with normal gland development (control group) and gland development inhibited by progesterone (experimental group). Animals were sacrificed on the PN Days 5, 10 and 15. YAP and p-YAP by immunohistochemistry and immunoblotting techniques to identify the main components of Hippo Signaling Pathway. YAP, LATS1, LATS 2, MST1, NF2 and TAZ used for the RT-qPCR methods. In conclusion, Hippo signaling pathway components were reduced during the adenogenesis process in mouse with impaired gland development.
Collapse
Affiliation(s)
- İrem İnanç
- Faculty of Medicine, Department of Histology and Embryology, Ankara University, Ankara, Turkey.
| | - Onur Bender
- Biotechnology Institute, Ankara University, Ankara, Turkey
| | - Arzu Atalay
- Biotechnology Institute, Ankara University, Ankara, Turkey
| | - Serdal Kenan Köse
- Faculty of Medicine, Department of Biostatistics, Ankara University, Ankara, Turkey
| | - Esra Erdemli
- Faculty of Medicine, Department of Histology and Embryology, Ankara University, Ankara, Turkey
| |
Collapse
|
5
|
Guo Q, Qin H, Chen Z, Zhang W, Zheng L, Qin T. Key roles of ubiquitination in regulating critical regulators of cancer stem cell functionality. Genes Dis 2025; 12:101311. [PMID: 40034124 PMCID: PMC11875185 DOI: 10.1016/j.gendis.2024.101311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 01/23/2024] [Accepted: 03/07/2024] [Indexed: 03/05/2025] Open
Abstract
The ubiquitin (Ub) system, a ubiquitous presence across eukaryotes, plays a crucial role in the precise orchestration of diverse cellular protein processes. From steering cellular signaling pathways and orchestrating cell cycle progression to guiding receptor trafficking and modulating immune responses, this process plays a crucial role in regulating various biological functions. The dysregulation of Ub-mediated signaling pathways in prevalent cancers ushers in a spectrum of clinical outcomes ranging from tumorigenesis and metastasis to recurrence and drug resistance. Ubiquitination, a linchpin process mediated by Ub, assumes a central mantle in molding cellular signaling dynamics. It navigates transitions in biological cues and ultimately shapes the destiny of proteins. Recent years have witnessed an upsurge in the momentum surrounding the development of protein-based therapeutics aimed at targeting the Ub system under the sway of cancer stem cells. The article provides a comprehensive overview of the ongoing in-depth discussions regarding the regulation of the Ub system and its impact on the development of cancer stem cells. Amidst the tapestry of insights, the article delves into the expansive roles of E3 Ub ligases, deubiquitinases, and transcription factors entwined with cancer stem cells. Furthermore, the spotlight turns to the interplay with pivotal signaling pathways the Notch, Hedgehog, Wnt/β-catenin, and Hippo-YAP signaling pathways all play crucial roles in the regulation of cancer stem cells followed by the specific modulation of Ub-proteasome.
Collapse
Affiliation(s)
- Qianqian Guo
- Department of Pharmacy, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan 450008, China
| | - Hai Qin
- Department of Clinical Laboratory, Beijing Jishuitan Hospital Guizhou Hospital, Guiyang, Guizhou 550014, China
| | - Zelong Chen
- The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Artificial Intelligence and IoT Smart Medical Engineering Research Center of Henan Province, Zhengzhou, Henan 450008, China
| | - Wenzhou Zhang
- Department of Pharmacy, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan 450008, China
| | - Lufeng Zheng
- School of Life Science and Technology, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Tingting Qin
- Department of Pharmacy, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan 450008, China
| |
Collapse
|
6
|
Wu Y, Lee TH, Cheng OH, Peden EK, Li Q, Wang J, Huang F, Melancon MP, Sheikh-Hamad D, Wang T, Truong L, Mitch WE, Liang M, Cheng J. Interplay between Skeletal Muscle Catabolism and Remodeling of Arteriovenous Fistula by Yes-Associated Protein 1 (YAP1) Signaling. J Am Soc Nephrol 2025; 36:845-858. [PMID: 39883520 PMCID: PMC12059102 DOI: 10.1681/asn.0000000605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 01/13/2025] [Indexed: 01/31/2025] Open
Abstract
Key Points Atrophied muscle–derived myostatin stimulated mesenchymal stem cell differentiation and adverse arteriovenous (AV) fistula remodeling through yes-associated protein 1 (YAP1) activation. Treatment with myostatin peptibody inhibited muscle wasting and blocked mesenchymal stem cell activation and AV fistula fibrosis. A light-sensitive drug-release strategy was engineered for the periadventitial delivery of verteporfin to improve AV fistula patency. Background Arteriovenous (AV) fistulas are the preferred access for dialysis but have a high incidence of failure. The aim of this study was to understand the crosstalk between skeletal muscle catabolism and AV fistula maturation failure. Methods Skeletal muscle metabolism and AV fistula maturation were evaluated in mice with CKD. The roles of myostatin and yes-associated protein 1 (YAP1) in regulating the transdifferentiation of adventitial mesenchymal stem cells (MSCs) and intima hyperplasia in AV fistula were investigated. Nanoparticles carrying a YAP1 inhibitor, verteporfin, with light irradiation–controlled release were synthesized and applied to AV fistula. Results Increased trichrome signals and stenosis were observed in AV fistulas from mice treated with myostatin and from mice with CKD. By contrast, blocking myostatin function with an anti-myostatin peptibody not only improved body weight and muscle size in CKD mice but also decreased neointima formation in AV fistulas. In cultured MSCs, myostatin induced YAP1 expression, promoting the differentiation of MSCs into myofibroblasts and inducing extracellular matrix deposition. Red light irradiation–controlled release of verteporfin from nanoparticles blocked YAP1 activation and alleviated myostatin-induced MSC activation. Periadventitial application and red light irradiation of nanoparticles carrying verteporfin significantly suppressed stiffening and neointima formation in AV fistula. Conclusions CKD induced muscle wasting, leading to increased production of myostatin, which stimulated MSC activation and vascular fibrosis linked to AV fistula stenosis. YAP1 signaling was activated in these processes. Red light irradiation–controlled release of verteporfin offered a feasible approach for local vascular drug intervention to improve AV fistula maturation.
Collapse
Affiliation(s)
- Yongdong Wu
- Section of Nephrology, Department of Medicine, Selzman Institute for Kidney Health, Baylor College of Medicine, Houston, Texas
- Department of Nephrology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China
| | - Tae Hoon Lee
- Section of Nephrology, Department of Medicine, Selzman Institute for Kidney Health, Baylor College of Medicine, Houston, Texas
| | - Owen H. Cheng
- Section of Nephrology, Department of Medicine, Selzman Institute for Kidney Health, Baylor College of Medicine, Houston, Texas
| | - Eric K. Peden
- Department of Surgery, Houston Methodist Hospital, Houston, Texas
| | - Qingtian Li
- Section of Nephrology, Department of Medicine, Selzman Institute for Kidney Health, Baylor College of Medicine, Houston, Texas
| | - Jun Wang
- Department of Pediatrics, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas
| | - Fengzhang Huang
- Section of Nephrology, Department of Medicine, Selzman Institute for Kidney Health, Baylor College of Medicine, Houston, Texas
| | - Marites P. Melancon
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - David Sheikh-Hamad
- Section of Nephrology, Department of Medicine, Selzman Institute for Kidney Health, Baylor College of Medicine, Houston, Texas
| | - Tao Wang
- Section of Nephrology, Department of Medicine, Selzman Institute for Kidney Health, Baylor College of Medicine, Houston, Texas
| | - Luan Truong
- Department of Pathology and Genomic medicine, Houston Methodist Hospital, Houston, Texas
| | - William E. Mitch
- Section of Nephrology, Department of Medicine, Selzman Institute for Kidney Health, Baylor College of Medicine, Houston, Texas
| | - Ming Liang
- Department of Nephrology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China
| | - Jizhong Cheng
- Section of Nephrology, Department of Medicine, Selzman Institute for Kidney Health, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
7
|
Meinhardt G, Waldhäusl H, Lackner AI, Wächter J, Maxian T, Höbler AL, Vondra S, Kunihs V, Saleh L, Haslinger P, Kiraly P, Szilagyi A, Than NG, Pollheimer J, Haider S, Knöfler M. The multifaceted roles of the transcriptional coactivator TAZ in extravillous trophoblast development of the human placenta. Proc Natl Acad Sci U S A 2025; 122:e2426385122. [PMID: 40228123 PMCID: PMC12037006 DOI: 10.1073/pnas.2426385122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 03/07/2025] [Indexed: 04/16/2025] Open
Abstract
Insights into the molecular processes that drive early development of the human placenta is crucial for our understanding of pregnancy complications such as preeclampsia and fetal growth restriction, since defects in maturation of its epithelial cell, the trophoblast, have been detected in the severe forms of these diseases. However, key regulators specifying the differentiated trophoblast subtypes of the placenta are only slowly emerging. By using diverse trophoblast cell models, we herein show that the transcriptional coactivator of HIPPO signaling, TAZ, plays a pivotal role in the development of invasive extravillous trophoblasts (EVTs), cells that are essential for decidual vessel remodeling and adaption of maternal blood flow to the placenta. Ribonucleic acid sequencing (RNA-seq) or protein analyses upon TAZ gene silencing or CRISPR-Cas9-mediated knockout in differentiating trophoblast stem cells, organoids, primary EVTs, choriocarcinoma cells, or villous explant cultures unraveled that the coactivator promoted expression of genes associated with EVT identity, motility, and survival. Accordingly, depletion or chemical inhibition of TAZ, interacting with TEA domain family member 1 (TEAD1), impaired EVT differentiation, invasion, and migration and triggered apoptosis in the different trophoblast models. Notably, the coactivator also suppressed cell cycle genes and regulators of trophoblast self-renewal and prevented EVTs from cell fusion in organoids and primary cultures. Moreover, TAZ promoted human leukocyte antigen G (HLA-G) surface expression and increased NUAK1 kinase in EVTs thereby maintaining its own expression. In summary, the transcriptional coactivator TAZ plays a multifaceted role in the development of the EVT cell lineage by controlling different biological processes that initiate and preserve differentiation.
Collapse
Affiliation(s)
- Gudrun Meinhardt
- Placental Development Group, Reproductive Biology Unit, Department of Obstetrics and Gynaecology, Medical University of Vienna, ViennaA-1090, Austria
| | - Hanna Waldhäusl
- Placental Development Group, Reproductive Biology Unit, Department of Obstetrics and Gynaecology, Medical University of Vienna, ViennaA-1090, Austria
| | - Andreas I. Lackner
- Maternal-Fetal Immunology Group, Reproductive Biology Unit, Department of Obstetrics and Gynaecology, Medical University of Vienna, ViennaA-1090, Austria
| | - Jasmin Wächter
- Placental Development Group, Reproductive Biology Unit, Department of Obstetrics and Gynaecology, Medical University of Vienna, ViennaA-1090, Austria
| | - Theresa Maxian
- Placental Development Group, Reproductive Biology Unit, Department of Obstetrics and Gynaecology, Medical University of Vienna, ViennaA-1090, Austria
| | - Anna-Lena Höbler
- Maternal-Fetal Immunology Group, Reproductive Biology Unit, Department of Obstetrics and Gynaecology, Medical University of Vienna, ViennaA-1090, Austria
| | - Sigrid Vondra
- Maternal-Fetal Immunology Group, Reproductive Biology Unit, Department of Obstetrics and Gynaecology, Medical University of Vienna, ViennaA-1090, Austria
| | - Victoria Kunihs
- Placental Development Group, Reproductive Biology Unit, Department of Obstetrics and Gynaecology, Medical University of Vienna, ViennaA-1090, Austria
| | - Leila Saleh
- Placental Development Group, Reproductive Biology Unit, Department of Obstetrics and Gynaecology, Medical University of Vienna, ViennaA-1090, Austria
| | - Peter Haslinger
- Maternal-Fetal Immunology Group, Reproductive Biology Unit, Department of Obstetrics and Gynaecology, Medical University of Vienna, ViennaA-1090, Austria
| | - Peter Kiraly
- Systems Biology of Reproduction Lendulet Group, Institute of Molecular Life Sciences, Hungarian Research Network (HUN-REN) Research Centre for Natural Sciences, Budapest1117, Hungary
| | - Andras Szilagyi
- Systems Biology of Reproduction Lendulet Group, Institute of Molecular Life Sciences, Hungarian Research Network (HUN-REN) Research Centre for Natural Sciences, Budapest1117, Hungary
| | - Nandor G. Than
- Systems Biology of Reproduction Lendulet Group, Institute of Molecular Life Sciences, Hungarian Research Network (HUN-REN) Research Centre for Natural Sciences, Budapest1117, Hungary
- Maternity Private Clinic of Obstetrics and Gynecology, Budapest1126, Hungary
- Department of Obstetrics and Gynecology, Semmelweis University, Budapest1088, Hungary
| | - Jürgen Pollheimer
- Maternal-Fetal Immunology Group, Reproductive Biology Unit, Department of Obstetrics and Gynaecology, Medical University of Vienna, ViennaA-1090, Austria
| | - Sandra Haider
- Placental Development Group, Reproductive Biology Unit, Department of Obstetrics and Gynaecology, Medical University of Vienna, ViennaA-1090, Austria
| | - Martin Knöfler
- Placental Development Group, Reproductive Biology Unit, Department of Obstetrics and Gynaecology, Medical University of Vienna, ViennaA-1090, Austria
| |
Collapse
|
8
|
Whited A, Elkhalil A, Clark G, Ghose P. CDH-3/Cadherin, YAP-1/YAP and EGL-44/TEAD promote SYX-2/Syntaxin and EFF-1 fusogen-mediated phagosome closure. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.02.646655. [PMID: 40236144 PMCID: PMC11996554 DOI: 10.1101/2025.04.02.646655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Physical interactions between cells, such as cell-cell junctions, can profoundly impact cell fate. A vital cell fate for normal development and homeostasis is programmed cell death. Cells fated to die must be efficiently cleared away via phagocytosis, and defects are associated with a variety of diseased states. Whether cell-cell physical associations affect programmed cell elimination has not been well-explored. Here we describe, in vivo, a cell-cell adhesion-driven signaling pathway that ensures compartment-specific cell clearance during development. We previously described the specialized cell death program "Compartmentalized Cell Elimination" (CCE) in the C. elegans embryo. During CCE, the tail-spike cell (TSC), a polarized epithelial cell, undergoes a tripartite, ordered, and organized death sequence, allowing for the study of three distinct death modalities in a single cell setting. Prior to its demise, the TSC serves as a scaffold for the tail tip, formed by the hyp10 epithelial cell which develops along the TSC process. The hyp10 cell in turn also serves as the phagocyte for the dying TSC process. Here we present data suggesting that the physical association between the dying TSC and hyp10 phagocyte via CDH-3/cadherin mediates function of the mechanosensitive transcriptional coactivator YAP-1/YAP and its partner EGL-44/TEAD in the hyp10 phagocyte to promote localization of hyp10 SYX-2/Syntaxin around the dying TSC remnant. This pathway facilitates the phagocytic function of EFF-1/fusogen, which we have previously shown to be required for phagosome sealing during CCE. Our work sheds additional light on a poorly understood step of phagocytosis and implicates adhesive forces and signaling between cells as important in cell uptake.
Collapse
|
9
|
Haddad A, Golan-Lev T, Benvenisty N, Goldberg M. Genome-wide screening in human embryonic stem cells identifies genes and pathways involved in the p53 pathway. Mol Med 2025; 31:97. [PMID: 40082762 PMCID: PMC11907909 DOI: 10.1186/s10020-025-01141-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 02/25/2025] [Indexed: 03/16/2025] Open
Abstract
BACKGROUND The tumor suppressor protein, p53, which is mutated in half of human tumors, plays a critical role in cellular responses to DNA damage and maintenance of genome stability. Therefore, increasing our understanding of the p53 pathway is essential for improving cancer treatment and diagnosis. METHODS This study, which aimed to identify genes and pathways that mediate resistance to p53 upregulation, used genome-wide CRISPR-Cas9 loss-of-function screening done with Nutlin-3a, which inhibits p53-MDM2 interaction, resulting in p53 accumulation and apoptotic cell death. We used bioinformatics analysis for the identification of genes and pathways that are involved in the p53 pathway and cell survival assays to validate specific genes. In addition, we used RNA-seq to identify differentially expressed p53 target genes in gene knockout (KO) cell lines. RESULTS Our screen revealed three significantly enriched pathways: The heparan sulfate glycosaminoglycan biosynthesis, diphthamide biosynthesis and Hippo pathway. Notably, TRIP12 was significantly enriched in our screen. We found that TRIP12 is required for the p53-dependent transcription of several pro-apoptotic genes. CONCLUSION Our study has identified two novel pathways that play a role in p53-mediated growth restriction. Moreover, we have highlighted the interaction between the Hippo and the p53 pathways. Interestingly, we have shown that TRIP12 plays an important function in the p53 pathway by selectively affecting its role as a transcription factor.
Collapse
Affiliation(s)
- Amir Haddad
- Department of Genetics, The Alexander Silberman Institute of Life Sciences, the Hebrew University of Jerusalem, 91904, Jerusalem, Israel
- The Azrieli Center for Stem Cells and Genetic Research, The Alexander Silberman Institute of Life Sciences, the Hebrew University of Jerusalem, 91904, Jerusalem, Israel
| | - Tamar Golan-Lev
- Department of Genetics, The Alexander Silberman Institute of Life Sciences, the Hebrew University of Jerusalem, 91904, Jerusalem, Israel
- The Azrieli Center for Stem Cells and Genetic Research, The Alexander Silberman Institute of Life Sciences, the Hebrew University of Jerusalem, 91904, Jerusalem, Israel
| | - Nissim Benvenisty
- Department of Genetics, The Alexander Silberman Institute of Life Sciences, the Hebrew University of Jerusalem, 91904, Jerusalem, Israel
- The Azrieli Center for Stem Cells and Genetic Research, The Alexander Silberman Institute of Life Sciences, the Hebrew University of Jerusalem, 91904, Jerusalem, Israel
| | - Michal Goldberg
- Department of Genetics, The Alexander Silberman Institute of Life Sciences, the Hebrew University of Jerusalem, 91904, Jerusalem, Israel.
| |
Collapse
|
10
|
Dicipulo R, Selland LG, Carpenter RG, Waskiewicz AJ. Functional role for Taz during hindbrain ventricle morphogenesis. PLoS One 2025; 20:e0313262. [PMID: 40080483 PMCID: PMC11906067 DOI: 10.1371/journal.pone.0313262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 10/21/2024] [Indexed: 03/15/2025] Open
Abstract
The brain ventricle system, composed of the ventricular cavities and the cerebral spinal fluid within, performs critical functions including circulation of nutrients, removal of wastes, and cushioning of neural tissues. Development of the hindbrain ventricle requires a series of factors that coordinate its initial formation and subsequent inflation. Previous work has demonstrated that the transcriptional co-activator Taz (also known as WW domain-containing transcription regulator protein 1, Wwtr1), a component of Hippo signalling, is active at hindbrain rhombomere boundaries where it is regulated by mechanotransduction and promotes proliferation. Here, we demonstrate that Taz is also a critical regulator of hindbrain ventricle development. Zebrafish embryos that lack Taz protein fail to undergo initial midline separation of the hindbrain ventricle. Furthermore, the ventricle phenotype is a result of disorganized cytoskeletal F-actin and apicobasal polarity components. In addition, we have demonstrated that the hindbrain rhombomere boundaries are a location of active Wnt-Hippo crosstalk. Through our work, we propose a model where Taz protein is stabilized at rhombomere boundaries and promotes proper cell polarity necessary for formation of the brain ventricle.
Collapse
Affiliation(s)
- Renée Dicipulo
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Lyndsay G. Selland
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Rowan G. Carpenter
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Andrew J. Waskiewicz
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
- Women & Children’s Health Research Institute, University of Alberta, Edmonton, Canada
| |
Collapse
|
11
|
Yuan Q, Yuan Y, Peng Y, Xia X, Chen Q, Yu FX, Feng X. Distinct effects of Hippo-YAP/TAZ and YAP/TAZ-TEAD in epithelial maintenance and repair. Biochem Biophys Res Commun 2025; 751:151427. [PMID: 39903968 DOI: 10.1016/j.bbrc.2025.151427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Accepted: 01/29/2025] [Indexed: 02/06/2025]
Abstract
The maintenance of epithelial homeostasis is essential for preserving tissue architecture and function, and the transcriptional co-activators YAP/TAZ are central to this regulatory network. Although the Hippo-YAP/TAZ-TEAD axis is known to govern epithelial integrity, it remains unclear to what extent Hippo-controlled YAP/TAZ activity overlaps with, or diverges from, YAP/TAZ-TEAD-dependent transcriptional programs in maintaining epithelial homeostasis. Here, we address this question by employing two complementary mouse models: "SuperHippo," which suppresses YAP/TAZ activity through enhanced Hippo pathway engagement, and "TEADi," which selectively disrupts YAP/TAZ-TEAD interactions. Our results revealed that while both models led to increased epithelial thickness in skin epithelial, SuperHippo mice exhibited pronounced epithelial impairment in oral mucosa, and markedly delayed wound healing. In contrast, TEADi mice displayed tissue-specific phenotypes with minimal disruption to oral epithelium integrity or wound repair. These findings indicate that Hippo-mediated YAP/TAZ regulation may extend beyond TEAD-dependent transcription. Our work clarifies the distinct contributions of Hippo-YAP/TAZ signaling and YAP/TAZ-TEAD interaction to epithelial maintenance and provides a basis for the development of therapeutic strategies targeting YAP/TAZ in epithelial disorders.
Collapse
Affiliation(s)
- Qiuyun Yuan
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yao Yuan
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China.
| | - Yang Peng
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Xiaoqiang Xia
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Qianming Chen
- Key Laboratory of Oral Biomedical Research of Zhejiang Province, Affiliated Stomatology Hospital, Zhejiang University School of Stomatology, Hangzhou, 310006, Zhejiang, China
| | - Fa-Xing Yu
- Institute of Pediatrics, Children's Hospital of Fudan University, Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Xiaodong Feng
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
12
|
Zhang Q, Yuan Y, Wang B, Gong P, Xiang L. Lysophosphatidic acid regulates implant osseointegration in murine models via YAP. Connect Tissue Res 2025; 66:87-95. [PMID: 39902934 DOI: 10.1080/03008207.2025.2459856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 01/23/2025] [Indexed: 02/06/2025]
Abstract
BACKGROUND Lysophosphatidic acid (LPA), a simple bioactive lysophospholipid, has been reported to regulate bone homeostasis and bone remodeling. This study aimed to elucidate the function and intrinsic mechanism of LPA in osseointegration in murine models. METHOD We constructed immediate implant models in murine maxillae. Micro-CT, H&E staining, and PCR assays were performed to evaluate the effects of LPA on osseointegration. Furthermore, Prx1-Cre;Yapf/f mice and Sp7-Cre;Yapf/f mice were generated to investigate the role of YAP on LPA-induced osseointegration. RESULT In this study, we identified that LPA might promote bone deposition on the tissue-implant interface and improve osseointegration. In addition, conditional knockout of YAP from MCSs and pre-osteoblasts blunts LPA-induced osteogenesis and osseointegration in mice. CONCLUSION Our data demonstrated that LPA-YAP signaling is particularly important to regulate osseointegration, which expands our understanding of LPA and provide the potential of LPA to be used in osseointegration.
Collapse
Affiliation(s)
- Qin Zhang
- Department of Oral Implantology, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Institute of Stomatology, Nanjing University, Nanjing, China
| | - Ying Yuan
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Bin Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ping Gong
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Lin Xiang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
13
|
Shan AP, Hlaing KS, Leelahavanichkul A, Amornphimoltham P. Efficacy of metformin and verteporfin treatment alone or in combination in a murine head and neck cancer xenograft model. Eur J Oral Sci 2025; 133:e13034. [PMID: 39780329 DOI: 10.1111/eos.13034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025]
Abstract
Despite treatment advances, head and neck squamous cell carcinoma (HNSCC) still poses a significant global health challenge. Combination therapies have emerged as more effective strategies than traditional chemotherapy in clinical practice by improving tumor response rates and patient survival while minimizing treatment-related toxicity. This study investigates the anticancer effects of metformin and verteporfin (Yes-associated protein 1 [YAP1] inhibitor) alone or in combination in HNSCC using vitro and in vivo approaches. Quantitative RT-PCR analysis of HNSCC cell lines reveals upregulation of YAP1 and related genes in the Hippo signaling pathway. Cell viability assays demonstrate a beneficial synergistic effect between metformin and verteporfin in inhibiting HNSCC tumor growth. In male BALB/cAJcl-nu/nu mice harboring HNSCC tumor xenografts, intraperitoneal administration of metformin and verteporfin enhances the inhibitory effect on tumor growth and suppresses YAP1 nuclear translocation when compared to vehicle or monotherapies. Furthermore, combination of these drugs reduces tumor cell proliferation (marked by Ki-67) and inhibits phosphoS6 ribosomal protein, as observed through immunofluorescent and immunohistochemical (IHC) analyses. The in vivo study underscores the therapeutic potential of the dual targeting approach with metformin and verteporfin in treating HNSCC, with minimal toxicity.
Collapse
Affiliation(s)
- Aung Phyo Shan
- Department of Oral Bioscience and Dental Public Health, International College of Dentistry, Walailak University, Bangkok, Thailand
| | - Khin Swe Hlaing
- Department of Oral Bioscience and Dental Public Health, International College of Dentistry, Walailak University, Bangkok, Thailand
| | - Asada Leelahavanichkul
- Translational Research in Inflammation and Immunology Research Unit, Department of Microbiology, Faculty of Medicine Chulalongkorn University, Bangkok, Thailand
| | - Panomwat Amornphimoltham
- Department of Oral Bioscience and Dental Public Health, International College of Dentistry, Walailak University, Bangkok, Thailand
- Department of Oral Biology, Faculty of Dentistry, Mahidol University, Bangkok, Thailand
| |
Collapse
|
14
|
Ferronato GDA, Rosa PMDS, Bridi A, Santos ACD, Nociti RP, Chiaratti MR, Perecin F, Meirelles FV, Sangalli JR, Silveira JCD. Transcriptomic effects of alginate hydrogel applied to the production of bovine embryos. Heliyon 2024; 10:e40957. [PMID: 39759294 PMCID: PMC11700250 DOI: 10.1016/j.heliyon.2024.e40957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 12/02/2024] [Accepted: 12/04/2024] [Indexed: 01/07/2025] Open
Abstract
In vitro-produced blastocysts are exposed to different stimuli when compared with in vivo ones. This includes the culture of in vitro embryos in a sturdy petri-dish, while in vivo embryos develop in a soft and dynamic structure. Here we hypothesized that a softer environment could differently modulate the in vitro produced embryos. To that aim, presumptive zygotes were produced by in vitro fertilization and divided into three groups: 1) Cultured in a regular Petri dish - Control (CON); 2) Cultured on top of an alginate hydrogel surface (TOP); 3) Encapsulated inside an alginate hydrogel sphere (ENC) and cultured. We observed a decrease in blastocyst rate in TOP and ENC compared with the CON. Profiling of 383 bovine miRNAs, we found 3 miRNAs involved in cell proliferation being differently modulated by the TOP and ENC groups (miR-1246; miR-1260b, and miR-541). Analyzing global levels of DNA methylation and hydroxymethylation, we observed increased levels of the two marks in the TOP group when compared with the CON and ENC systems. RNA sequencing (RNA-seq) analysis carried out using blastocysts showed alterations in several developmentally important genes among the three groups. In summary, our results indicate that in vitro embryo production was possible to achieve up to the blastocyst stage. However, with the experimental conditions used herein, the alginate hydrogels adversely affected the embryo development, which were paralleled by epigenetic and transcriptomic changes.
Collapse
Affiliation(s)
- Giuliana de A. Ferronato
- Faculty of Animal Science and Food Engineering, Department of Veterinary Medicine, University of São Paulo, Pirassununga, SP, Brazil
| | - Paola M. da S. Rosa
- Faculty of Animal Science and Food Engineering, Department of Veterinary Medicine, University of São Paulo, Pirassununga, SP, Brazil
| | - Alessandra Bridi
- Faculty of Animal Science and Food Engineering, Department of Veterinary Medicine, University of São Paulo, Pirassununga, SP, Brazil
| | - Angélica Camargo dos Santos
- Universidade Federal de São Carlos, Centro de Ciências Biológicas e da Saúde, Departamento de Genética e Evolução, São Carlos, SP, Brazil
| | - Ricardo P. Nociti
- Faculty of Animal Science and Food Engineering, Department of Veterinary Medicine, University of São Paulo, Pirassununga, SP, Brazil
| | - Marcos Roberto Chiaratti
- Universidade Federal de São Carlos, Centro de Ciências Biológicas e da Saúde, Departamento de Genética e Evolução, São Carlos, SP, Brazil
| | - Felipe Perecin
- Faculty of Animal Science and Food Engineering, Department of Veterinary Medicine, University of São Paulo, Pirassununga, SP, Brazil
| | - Flávio V. Meirelles
- Faculty of Animal Science and Food Engineering, Department of Veterinary Medicine, University of São Paulo, Pirassununga, SP, Brazil
| | - Juliano R. Sangalli
- Faculty of Animal Science and Food Engineering, Department of Veterinary Medicine, University of São Paulo, Pirassununga, SP, Brazil
| | - Juliano C. da Silveira
- Faculty of Animal Science and Food Engineering, Department of Veterinary Medicine, University of São Paulo, Pirassununga, SP, Brazil
| |
Collapse
|
15
|
Jafarinia H, Shi L, Wolfenson H, Carlier A. YAP phosphorylation within integrin adhesions: Insights from a computational model. Biophys J 2024; 123:3658-3668. [PMID: 39233443 PMCID: PMC11560305 DOI: 10.1016/j.bpj.2024.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 08/13/2024] [Accepted: 09/03/2024] [Indexed: 09/06/2024] Open
Abstract
Mechanical and biochemical cues intricately activate Yes-associated protein (YAP), which is pivotal for the cellular responses to these stimuli. Recent findings reveal an unexplored role of YAP in influencing the apoptotic process. It has been shown that, on soft matrices, YAP is recruited to small adhesions, phosphorylated at Y357, and translocated into the nucleus triggering apoptosis. Interestingly, YAP Y357 phosphorylation is significantly reduced in larger mature focal adhesions on stiff matrices. Building upon these novel insights, we have developed a stochastic model to delve deeper into the complex dynamics of YAP phosphorylation within integrin adhesions. Our findings emphasize several key points: firstly, increasing the cytosolic diffusion rate of YAP correlates with higher levels of phosphorylated YAP (pYAP); secondly, increasing the number of binding sites and distributing them across the membrane surface, mimicking smaller adhesions, leads to higher pYAP levels, particularly at lower diffusion rates. Moreover, we show that the binding and release rate of YAP to adhesions as well as adhesion lifetimes significantly influence the size effect of adhesion-induced YAP phosphorylation. The results highlight the complex and dynamic interplay between adhesion lifetime, the rate of pYAP unbinding from adhesions, and dephosphorylation rates, collectively shaping overall pYAP levels. In summary, our work advances the understanding of YAP mechanotransduction and opens avenues for experimental validation.
Collapse
Affiliation(s)
- Hamidreza Jafarinia
- MERLN Institute for Technology-Inspired Regenerative Medicine, Department of Cell Biology-Inspired Tissue Engineering, Maastricht University, Maastricht, the Netherlands
| | - Lidan Shi
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Haguy Wolfenson
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Aurélie Carlier
- MERLN Institute for Technology-Inspired Regenerative Medicine, Department of Cell Biology-Inspired Tissue Engineering, Maastricht University, Maastricht, the Netherlands.
| |
Collapse
|
16
|
Guijarro C, Song S, Aigouy B, Clément R, Villoutreix P, Kelly RG. Single-cell morphometrics reveals T-box gene-dependent patterns of epithelial tension in the Second Heart field. Nat Commun 2024; 15:9512. [PMID: 39496595 PMCID: PMC11535409 DOI: 10.1038/s41467-024-53612-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 10/17/2024] [Indexed: 11/06/2024] Open
Abstract
The vertebrate heart tube extends by progressive addition of epithelial second heart field (SHF) progenitor cells from the dorsal pericardial wall. The interplay between epithelial mechanics and genetic mechanisms during SHF deployment is unknown. Here, we present a quantitative single-cell morphometric analysis of SHF cells during heart tube extension, including force inference analysis of epithelial stress. Joint spatial Principal Component Analysis reveals that cell orientation and stress direction are the main parameters defining apical cell morphology and distinguishes cells adjacent to the arterial and venous poles. Cell shape and mechanical forces display a dynamic relationship during heart tube formation. Moreover, while the T-box transcription factor Tbx1 is necessary for cell orientation towards the arterial pole, activation of Tbx5 in the posterior SHF correlates with the establishment of epithelial stress and SHF deletion of Tbx5 relaxes the progenitor epithelium. Integrating findings from cell-scale feature patterning and mechanical stress provides new insights into cardiac morphogenesis.
Collapse
Affiliation(s)
- Clara Guijarro
- Aix-Marseille Université, CNRS UMR 7288, IBDM, Turing Centre for Living Systems, Marseille, France
- Aix-Marseille Université, LIS, UMR 7020, Turing Centre for Living Systems, Marseille, France
- Aix-Marseille Université, MMG, Inserm U1251, Turing Centre for Living Systems, Marseille, France
| | - Solène Song
- Aix-Marseille Université, LIS, UMR 7020, Turing Centre for Living Systems, Marseille, France
- Aix-Marseille Université, MMG, Inserm U1251, Turing Centre for Living Systems, Marseille, France
| | - Benoit Aigouy
- Aix-Marseille Université, CNRS UMR 7288, IBDM, Turing Centre for Living Systems, Marseille, France
| | - Raphaël Clément
- Aix-Marseille Université, CNRS UMR 7288, IBDM, Turing Centre for Living Systems, Marseille, France
| | - Paul Villoutreix
- Aix-Marseille Université, LIS, UMR 7020, Turing Centre for Living Systems, Marseille, France.
- Aix-Marseille Université, MMG, Inserm U1251, Turing Centre for Living Systems, Marseille, France.
| | - Robert G Kelly
- Aix-Marseille Université, CNRS UMR 7288, IBDM, Turing Centre for Living Systems, Marseille, France.
| |
Collapse
|
17
|
Lei Y, Liu Q, Chen B, Wu F, Li Y, Dong X, Ma N, Wu Z, Zhu Y, Wang L, Fu Y, Liu Y, Song Y, Du M, Zhang H, Zhu J, Lyons TJ, Wang T, Hu J, Xu H, Chen M, Yan H, Wang X. Protein O-GlcNAcylation coupled to Hippo signaling drives vascular dysfunction in diabetic retinopathy. Nat Commun 2024; 15:9334. [PMID: 39472558 PMCID: PMC11522279 DOI: 10.1038/s41467-024-53601-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 10/17/2024] [Indexed: 11/02/2024] Open
Abstract
Metabolic disorder significantly contributes to diabetic vascular complications, including diabetic retinopathy, the leading cause of blindness in the working-age population. However, the molecular mechanisms by which disturbed metabolic homeostasis causes vascular dysfunction in diabetic retinopathy remain unclear. O-GlcNAcylation modification acts as a nutrient sensor particularly sensitive to ambient glucose. Here, we observe pronounced O-GlcNAc elevation in retina endothelial cells of diabetic retinopathy patients and mouse models. Endothelial-specific depletion or pharmacological inhibition of O-GlcNAc transferase effectively mitigates vascular dysfunction. Mechanistically, we find that Yes-associated protein (YAP) and Transcriptional co-activator with PDZ-binding motif (TAZ), key effectors of the Hippo pathway, are O-GlcNAcylated in diabetic retinopathy. We identify threonine 383 as an O-GlcNAc site on YAP, which inhibits its phosphorylation at serine 397, leading to its stabilization and activation, thereby promoting vascular dysfunction by inducing a pro-angiogenic and glucose metabolic transcriptional program. This work emphasizes the critical role of the O-GlcNAc-Hippo axis in the pathogenesis of diabetic retinopathy and suggests its potential as a therapeutic target.
Collapse
Affiliation(s)
- Yi Lei
- Department of Ophthalmology, Laboratory of Molecular Ophthalmology and Tianjin Key Laboratory of Ocular Trauma, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Medical University General Hospital, Tianjin, China
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, State Key Laboratory of Experimental Hematology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Qiangyun Liu
- Department of Ophthalmology, Laboratory of Molecular Ophthalmology and Tianjin Key Laboratory of Ocular Trauma, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Medical University General Hospital, Tianjin, China
| | - Binggui Chen
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, State Key Laboratory of Experimental Hematology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Fangfang Wu
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, State Key Laboratory of Experimental Hematology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yiming Li
- Department of Ophthalmology, Laboratory of Molecular Ophthalmology and Tianjin Key Laboratory of Ocular Trauma, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Medical University General Hospital, Tianjin, China
| | - Xue Dong
- Department of Ophthalmology, Laboratory of Molecular Ophthalmology and Tianjin Key Laboratory of Ocular Trauma, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Medical University General Hospital, Tianjin, China
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, State Key Laboratory of Experimental Hematology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Nina Ma
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, State Key Laboratory of Experimental Hematology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Ziru Wu
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, State Key Laboratory of Experimental Hematology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yanfang Zhu
- Department of Ophthalmology, Laboratory of Molecular Ophthalmology and Tianjin Key Laboratory of Ocular Trauma, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Medical University General Hospital, Tianjin, China
| | - Lu Wang
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, State Key Laboratory of Experimental Hematology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yuxin Fu
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, State Key Laboratory of Experimental Hematology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yuming Liu
- Department of Ophthalmology, Laboratory of Molecular Ophthalmology and Tianjin Key Laboratory of Ocular Trauma, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Medical University General Hospital, Tianjin, China
| | - Yinting Song
- Department of Ophthalmology, Laboratory of Molecular Ophthalmology and Tianjin Key Laboratory of Ocular Trauma, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Medical University General Hospital, Tianjin, China
| | - Mei Du
- Department of Ophthalmology, Laboratory of Molecular Ophthalmology and Tianjin Key Laboratory of Ocular Trauma, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Medical University General Hospital, Tianjin, China
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, State Key Laboratory of Experimental Hematology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Heng Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Jidong Zhu
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, State Key Laboratory of Experimental Hematology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Timothy J Lyons
- Division of Endocrinology, Diabetes and Metabolic Diseases at the Medical University of South Carolina, Charleston, SC, USA
| | - Ting Wang
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, State Key Laboratory of Experimental Hematology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Junhao Hu
- Laboratory of Vascular Biology and Organ Homeostasis, Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Heping Xu
- The Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, UK
| | - Mei Chen
- The Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, UK
| | - Hua Yan
- Department of Ophthalmology, Laboratory of Molecular Ophthalmology and Tianjin Key Laboratory of Ocular Trauma, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Medical University General Hospital, Tianjin, China.
- School of Medicine, Nankai University, Tianjin, China.
| | - Xiaohong Wang
- Department of Ophthalmology, Laboratory of Molecular Ophthalmology and Tianjin Key Laboratory of Ocular Trauma, Ministry of Education International Joint Laboratory of Ocular Diseases, Tianjin Medical University General Hospital, Tianjin, China.
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, State Key Laboratory of Experimental Hematology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China.
| |
Collapse
|
18
|
Jahn J, Ehlen QT, Kaplan L, Best TM, Meng Z, Huang CY. Interplay of Glucose Metabolism and Hippo Pathway in Chondrocytes: Pathophysiology and Therapeutic Targets. Bioengineering (Basel) 2024; 11:972. [PMID: 39451348 PMCID: PMC11505586 DOI: 10.3390/bioengineering11100972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 09/24/2024] [Accepted: 09/24/2024] [Indexed: 10/26/2024] Open
Abstract
In this review, we explore the intricate relationship between glucose metabolism and mechanotransduction pathways, with a specific focus on the role of the Hippo signaling pathway in chondrocyte pathophysiology. Glucose metabolism is a vital element in maintaining proper chondrocyte function, but it has also been implicated in the pathogenesis of osteoarthritis (OA) via the induction of pro-inflammatory signaling pathways and the establishment of an intracellular environment conducive to OA. Alternatively, mechanotransduction pathways such as the Hippo pathway possess the capacity to respond to mechanical stimuli and have an integral role in maintaining chondrocyte homeostasis. However, these mechanotransduction pathways can be dysregulated and potentially contribute to the progression of OA. We discussed how alterations in glucose levels may modulate the Hippo pathway components via a variety of mechanisms. Characterizing the interaction between glucose metabolism and the Hippo pathway highlights the necessity of balancing both metabolic and mechanical signaling to maintain chondrocyte health and optimal functionality. Furthermore, this review demonstrates the scarcity of the literature on the relationship between glucose metabolism and mechanotransduction and provides a summary of current research dedicated to this specific area of study. Ultimately, increased research into this topic may elucidate novel mechanisms and relationships integrating mechanotransduction and glucose metabolism. Through this review we hope to inspire future research into this topic to develop innovative treatments for addressing the clinical challenges of OA.
Collapse
Affiliation(s)
- Jacob Jahn
- University of Miami Miller School of Medicine, Miami, FL 33136, USA; (J.J.); (Q.T.E.); (L.K.); (T.M.B.); (Z.M.)
| | - Quinn T. Ehlen
- University of Miami Miller School of Medicine, Miami, FL 33136, USA; (J.J.); (Q.T.E.); (L.K.); (T.M.B.); (Z.M.)
| | - Lee Kaplan
- University of Miami Miller School of Medicine, Miami, FL 33136, USA; (J.J.); (Q.T.E.); (L.K.); (T.M.B.); (Z.M.)
- Department of Orthopedics, University of Miami, Miami, FL 33136, USA
- UHealth Sports Medicine Institute, University of Miami, Miami, FL 33136, USA
| | - Thomas M. Best
- University of Miami Miller School of Medicine, Miami, FL 33136, USA; (J.J.); (Q.T.E.); (L.K.); (T.M.B.); (Z.M.)
- Department of Orthopedics, University of Miami, Miami, FL 33136, USA
- UHealth Sports Medicine Institute, University of Miami, Miami, FL 33136, USA
| | - Zhipeng Meng
- University of Miami Miller School of Medicine, Miami, FL 33136, USA; (J.J.); (Q.T.E.); (L.K.); (T.M.B.); (Z.M.)
- Department of Molecular and Cellular Pharmacology, Miller School of Medicine, Miami, FL 33136, USA
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Chun-Yuh Huang
- UHealth Sports Medicine Institute, University of Miami, Miami, FL 33136, USA
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL 33146, USA
| |
Collapse
|
19
|
Long X, Yang Y, Zhou K. Sepsis induces the cardiomyocyte apoptosis and cardiac dysfunction through activation of YAP1/Serpine1/caspase-3 pathway. Open Med (Wars) 2024; 19:20241018. [PMID: 39308919 PMCID: PMC11416050 DOI: 10.1515/med-2024-1018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 07/14/2024] [Accepted: 07/23/2024] [Indexed: 09/25/2024] Open
Abstract
Background Sepsis triggers myocardial injury and dysfunction, leading to a high mortality rate in patients. Cardiomyocyte apoptosis plays a positive regulatory role in septic myocardial injury and dysfunction. However, the mechanism is unclear. Methods Bioinformatics analysis was used to identify differentially expressed genes in septic mice heart and validate key genes and pathways. The correlation of protein-protein and protein-pathway was analyzed. Sequentially, the cecal ligament and puncture (CLP) was used to induce septic mice, followed by Serpine1 inhibitor treatment. Finally, the regulatory relationship of Yes-associated protein1 (YAP1), Serpine1, and caspase-3 was verified in LPS-exposed mouse cardiomyocytes. Results Bioinformatic analysis found that Serpine1 expression is decreased in septic mice heart tissue and closely related to the HIPPO signaling pathway, while YAP1 is negatively correlated with apoptosis. In vivo, CLP induced a reduction of survival rate, cardiac dysfunction, and an increase in Serpine1 and Cleaved Caspase-3 expression, which could be reversed by a Serpine1 inhibitor. In vitro, LPS induced the mouse cardiomyocytes apoptosis, which could be reversed by Serpine1 inhibitor. Silencing YAP1 and Serpine1 reversed the LPS-induced increase in Serpine1 and Cleaved Caspase-3 expression, but silencing Serpine1 did not affect the LPS-induced YAP1 expression. Conclusion Sepsis induced mouse cardiomyocytes apoptosis and cardiac dysfunction through activation of YAP1/Serpine1/caspase-3 pathway.
Collapse
Affiliation(s)
- Xueyuan Long
- Department of Cardiovascular Medicine, Chongqing University Central Hospital, Chongqing, 400014, China
| | - Yanpeng Yang
- Department of Cardiovascular Medicine, Chongqing University Central Hospital, Chongqing, 400014, China
| | - Ke Zhou
- Department of Cardiovascular Medicine, Chongqing University Central Hospital, Chongqing, 400014, China
| |
Collapse
|
20
|
Crossey E, Carty S, Shao F, Henao-Vasquez J, Ysasi AB, Zeng M, Hinds A, Lo M, Tilston-Lunel A, Varelas X, Jones MR, Fine A. Influenza induces lung lymphangiogenesis independent of YAP/TAZ activity in lymphatic endothelial cells. Sci Rep 2024; 14:21324. [PMID: 39266641 PMCID: PMC11393066 DOI: 10.1038/s41598-024-72115-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 09/03/2024] [Indexed: 09/14/2024] Open
Abstract
The lymphatic system consists of a vessel network lined by specialized lymphatic endothelial cells (LECs) that are responsible for tissue fluid homeostasis and immune cell trafficking. The mechanisms for organ-specific LEC responses to environmental cues are not well understood. We found robust lymphangiogenesis during influenza A virus infection in the adult mouse lung. We show that the number of LECs increases twofold at 7 days post-influenza infection (dpi) and threefold at 21 dpi, and that lymphangiogenesis is preceded by lymphatic dilation. We also show that the expanded lymphatic network enhances fluid drainage to mediastinal lymph nodes. Using EdU labeling, we found that a significantly higher number of pulmonary LECs are proliferating at 7 dpi compared to LECs in homeostatic conditions. Lineage tracing during influenza indicates that new pulmonary LECs are derived from preexisting LECs rather than non-LEC progenitors. Lastly, using a conditional LEC-specific YAP/TAZ knockout model, we established that lymphangiogenesis, fluid transport and the immune response to influenza are independent of YAP/TAZ activity in LECs. These findings were unexpected, as they indicate that YAP/TAZ signaling is not crucial for these processes.
Collapse
Affiliation(s)
- Erin Crossey
- Division of Pulmonary, Allergy, Sleep and Critical Care, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, 72 East Concord St, R-304, Boston, MA, 02118, USA.
| | - Senegal Carty
- Division of Pulmonary, Allergy, Sleep and Critical Care, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, 72 East Concord St, R-304, Boston, MA, 02118, USA
| | - Fengzhi Shao
- Division of Pulmonary, Allergy, Sleep and Critical Care, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, 72 East Concord St, R-304, Boston, MA, 02118, USA
| | - Jhonatan Henao-Vasquez
- Division of Pulmonary, Allergy, Sleep and Critical Care, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, 72 East Concord St, R-304, Boston, MA, 02118, USA
| | - Alexandra B Ysasi
- Division of Pulmonary, Allergy, Sleep and Critical Care, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, 72 East Concord St, R-304, Boston, MA, 02118, USA
| | - Michelle Zeng
- Division of Pulmonary, Allergy, Sleep and Critical Care, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, 72 East Concord St, R-304, Boston, MA, 02118, USA
| | - Anne Hinds
- Division of Pulmonary, Allergy, Sleep and Critical Care, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, 72 East Concord St, R-304, Boston, MA, 02118, USA
| | - Ming Lo
- Department of Pathology and Laboratory Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- Comparative Pathology Laboratory, Boston University National Emerging and Infectious Disease Laboratories, Boston, MA, USA
| | - Andrew Tilston-Lunel
- Department of Biochemistry and Cell Biology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Xaralabos Varelas
- Department of Biochemistry and Cell Biology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Matthew R Jones
- Division of Pulmonary, Allergy, Sleep and Critical Care, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, 72 East Concord St, R-304, Boston, MA, 02118, USA
| | - Alan Fine
- Division of Pulmonary, Allergy, Sleep and Critical Care, Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, 72 East Concord St, R-304, Boston, MA, 02118, USA
| |
Collapse
|
21
|
Nelson KM, Ferrick BJ, Karimi H, Hatem CL, Gleghorn JP. A straightforward cell culture insert model to incorporate biochemical and biophysical stromal properties into transplacental transport studies. Placenta 2024:S0143-4004(24)00637-4. [PMID: 39266436 DOI: 10.1016/j.placenta.2024.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 07/19/2024] [Accepted: 09/04/2024] [Indexed: 09/14/2024]
Abstract
The placental extracellular matrix (ECM) dynamically remodels over pregnancy and in disease. How these changes impact placental barrier function is poorly understood as there are limited in vitro models of the placenta with a modifiable stromal compartment to mechanistically investigate these extracellular factors. We developed a straightforward method to incorporate uniform hydrogels into standard cell culture inserts for transplacental transport studies. Uniform polyacrylamide (PAA) gels were polymerized within cell culture inserts by (re)using the insert packaging to create a closed, controllable environmental chamber. PAA pre-polymer solution was added dropwise via a syringe to the cell culture insert and the atmosphere was purged with an inert gas. Transport and cell culture studies were conducted to validate the model. We successfully incorporated ECM-functionalized uniform PAA gels into cell culture inserts, enabling cell adhesion and monolayer formation. Imaging and analyte transport studies validated gel formation and expected mass transport results, and successful cell studies confirmed cell viability, stiffness-mediated YAP translocation, and that the model could be used in transplacental transport studies. Detailed methods and validation protocols are included. The incorporation of a PAA gel within a cell culture insert enables independent study of placental ECM biophysical and biochemical properties in the context of transplacental transport. These straightforward and low-cost methods to build three-dimensional cellular models are readily adoptable by the wider scientific community.
Collapse
Affiliation(s)
- Katherine M Nelson
- Department of Biomedical Engineering, University of Delaware, Newark, DE, 19716, USA
| | - Bryan J Ferrick
- Department of Biomedical Engineering, University of Delaware, Newark, DE, 19716, USA
| | - Hassan Karimi
- Department of Biomedical Engineering, University of Delaware, Newark, DE, 19716, USA
| | - Christine L Hatem
- Department of Biomedical Engineering, University of Delaware, Newark, DE, 19716, USA
| | - Jason P Gleghorn
- Department of Biomedical Engineering, University of Delaware, Newark, DE, 19716, USA.
| |
Collapse
|
22
|
Jafarinia H, Khalilimeybodi A, Barrasa-Fano J, Fraley SI, Rangamani P, Carlier A. Insights gained from computational modeling of YAP/TAZ signaling for cellular mechanotransduction. NPJ Syst Biol Appl 2024; 10:90. [PMID: 39147782 PMCID: PMC11327324 DOI: 10.1038/s41540-024-00414-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 07/27/2024] [Indexed: 08/17/2024] Open
Abstract
YAP/TAZ signaling pathway is regulated by a multiplicity of feedback loops, crosstalk with other pathways, and both mechanical and biochemical stimuli. Computational modeling serves as a powerful tool to unravel how these different factors can regulate YAP/TAZ, emphasizing biophysical modeling as an indispensable tool for deciphering mechanotransduction and its regulation of cell fate. We provide a critical review of the current state-of-the-art of computational models focused on YAP/TAZ signaling.
Collapse
Affiliation(s)
- Hamidreza Jafarinia
- MERLN Institute for Technology-Inspired Regenerative Medicine, Department of Cell Biology-Inspired Tissue Engineering, Maastricht University, Maastricht, The Netherlands
| | - Ali Khalilimeybodi
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, CA, 92093-0411, USA
| | - Jorge Barrasa-Fano
- Department of Mechanical Engineering, Biomechanics Section, KU Leuven, Leuven, Belgium
| | - Stephanie I Fraley
- Department of Bioengineering, University of California San Diego, La Jolla, CA, 92093-0411, USA
| | - Padmini Rangamani
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, CA, 92093-0411, USA.
| | - Aurélie Carlier
- MERLN Institute for Technology-Inspired Regenerative Medicine, Department of Cell Biology-Inspired Tissue Engineering, Maastricht University, Maastricht, The Netherlands.
| |
Collapse
|
23
|
Jiang L, Yan C, Yi Y, Zhu L, Liu Z, Zhang D, Jiang W. Cell size regulates human endoderm specification through actomyosin-dependent AMOT-YAP signaling. Stem Cell Reports 2024; 19:1137-1155. [PMID: 39094563 PMCID: PMC11368700 DOI: 10.1016/j.stemcr.2024.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 06/29/2024] [Accepted: 07/01/2024] [Indexed: 08/04/2024] Open
Abstract
Cell size is a crucial physical property that significantly impacts cellular physiology and function. However, the influence of cell size on stem cell specification remains largely unknown. Here, we investigated the dynamic changes in cell size during the differentiation of human pluripotent stem cells into definitive endoderm (DE). Interestingly, cell size exhibited a gradual decrease as DE differentiation progressed with higher stiffness. Furthermore, the application of hypertonic pressure or chemical to accelerate the reduction in cell size significantly and specifically enhanced DE differentiation. By functionally intervening in mechanosensitive elements, we have identified actomyosin activity as a crucial mediator of both DE differentiation and cell size reduction. Mechanistically, the reduction in cell size induces actomyosin-dependent angiomotin (AMOT) nuclear translocation, which suppresses Yes-associated protein (YAP) activity and thus facilitates DE differentiation. Together, our study has established a novel connection between cell size diminution and DE differentiation, which is mediated by AMOT nuclear translocation. Additionally, our findings suggest that the application of osmotic pressure can effectively promote human endodermal lineage differentiation.
Collapse
Affiliation(s)
- Lai Jiang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, Wuhan 430062, China; Department of Biological Repositories, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Chenchao Yan
- Department of Biological Repositories, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Ying Yi
- Department of Biological Repositories, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Lihang Zhu
- Department of Biological Repositories, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Zheng Liu
- The Institute for Advanced Studies, Wuhan University, Wuhan, China
| | - Donghui Zhang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, Wuhan 430062, China.
| | - Wei Jiang
- Department of Biological Repositories, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, China.
| |
Collapse
|
24
|
Zhu S, Chen W, Masson A, Li YP. Cell signaling and transcriptional regulation of osteoblast lineage commitment, differentiation, bone formation, and homeostasis. Cell Discov 2024; 10:71. [PMID: 38956429 PMCID: PMC11219878 DOI: 10.1038/s41421-024-00689-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 05/04/2024] [Indexed: 07/04/2024] Open
Abstract
The initiation of osteogenesis primarily occurs as mesenchymal stem cells undergo differentiation into osteoblasts. This differentiation process plays a crucial role in bone formation and homeostasis and is regulated by two intricate processes: cell signal transduction and transcriptional gene expression. Various essential cell signaling pathways, including Wnt, BMP, TGF-β, Hedgehog, PTH, FGF, Ephrin, Notch, Hippo, and Piezo1/2, play a critical role in facilitating osteoblast differentiation, bone formation, and bone homeostasis. Key transcriptional factors in this differentiation process include Runx2, Cbfβ, Runx1, Osterix, ATF4, SATB2, and TAZ/YAP. Furthermore, a diverse array of epigenetic factors also plays critical roles in osteoblast differentiation, bone formation, and homeostasis at the transcriptional level. This review provides an overview of the latest developments and current comprehension concerning the pathways of cell signaling, regulation of hormones, and transcriptional regulation of genes involved in the commitment and differentiation of osteoblast lineage, as well as in bone formation and maintenance of homeostasis. The paper also reviews epigenetic regulation of osteoblast differentiation via mechanisms, such as histone and DNA modifications. Additionally, we summarize the latest developments in osteoblast biology spurred by recent advancements in various modern technologies and bioinformatics. By synthesizing these insights into a comprehensive understanding of osteoblast differentiation, this review provides further clarification of the mechanisms underlying osteoblast lineage commitment, differentiation, and bone formation, and highlights potential new therapeutic applications for the treatment of bone diseases.
Collapse
Affiliation(s)
- Siyu Zhu
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA, USA
| | - Wei Chen
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA, USA.
| | - Alasdair Masson
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA, USA
| | - Yi-Ping Li
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA, USA.
| |
Collapse
|
25
|
Kiang KM, Ahad L, Zhong X, Lu QR. Biomolecular condensates: hubs of Hippo-YAP/TAZ signaling in cancer. Trends Cell Biol 2024; 34:566-577. [PMID: 38806345 DOI: 10.1016/j.tcb.2024.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 04/14/2024] [Accepted: 04/30/2024] [Indexed: 05/30/2024]
Abstract
Biomolecular condensates, the membraneless cellular compartments formed by liquid-liquid phase separation (LLPS), represent an important mechanism for physiological and tumorigenic processes. Recent studies have advanced our understanding of how these condensates formed in the cytoplasm or nucleus regulate Hippo signaling, a central player in organogenesis and tumorigenesis. Here, we review recent findings on the dynamic formation and function of biomolecular condensates in regulating the Hippo-yes-associated protein (YAP)/transcription coactivator with PDZ-binding motif (TAZ) signaling pathway under physiological and pathological processes. We further discuss how the nuclear condensates of YAP- or TAZ-fusion oncoproteins compartmentalize crucial transcriptional co-activators and alter chromatin architecture to promote oncogenic programs. Finally, we highlight key questions regarding how these findings may pave the way for novel therapeutics to target cancer.
Collapse
Affiliation(s)
- Karrie M Kiang
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Surgery, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - Leena Ahad
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Xiaowen Zhong
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Q Richard Lu
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| |
Collapse
|
26
|
Dai E, Wang W, Li Y, Ye D, Li Y. Lactate and lactylation: Behind the development of tumors. Cancer Lett 2024; 591:216896. [PMID: 38641309 DOI: 10.1016/j.canlet.2024.216896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/13/2024] [Accepted: 04/11/2024] [Indexed: 04/21/2024]
Abstract
There is growing evidence that lactate can have a wide range of biological impacts in addition to being a waste product of metabolism. Because of the Warburg effect, tumors generate lots of lactate, which create a tumor microenvironment (TME) with low nutrition, hypoxia, and low pH. As a result, the immunosuppressive network is established to gain immune escape potential and regulate tumor growth. Consequently, the tumor lactate pathway is emerging as a possible therapeutic target for tumor. Importantly, Zhao et al. first discovered histone lysine lactylation (Kla) in 2019, which links gene regulation to cell metabolism through dysmetabolic activity and epigenetic modifications, influencing TME and tumor development. Therefore, the aim of this paper is to explore the effects of lactate and lactylation on the TME and tumors, and provide theoretical basis for further research on potential therapeutic targets and biomarkers, with the view to providing new ideas and methods for tumor treatment and prognosis evaluation.
Collapse
Affiliation(s)
- Enci Dai
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 650 Xinsongjiang Road, Shanghai, 201600, China.
| | - Wei Wang
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 650 Xinsongjiang Road, Shanghai, 201600, China.
| | - Yingying Li
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 650 Xinsongjiang Road, Shanghai, 201600, China.
| | - Defeng Ye
- Department of Thoracic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No. 100 Haining Road, Hongkou District, Shanghai, 200080, China.
| | - Yanli Li
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 650 Xinsongjiang Road, Shanghai, 201600, China.
| |
Collapse
|
27
|
Kim G, Bhattarai PY, Lim SC, Lee KY, Choi HS. Sirtuin 5-mediated deacetylation of TAZ at K54 promotes melanoma development. Cell Oncol (Dordr) 2024; 47:967-985. [PMID: 38112979 DOI: 10.1007/s13402-023-00910-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/04/2023] [Indexed: 12/21/2023] Open
Abstract
PURPOSE Nuclear accumulation of YAP/TAZ promotes tumorigenesis in several cancers, including melanoma. Although the mechanisms underlying the nuclear retention of YAP are known, those underlying the retention of TAZ remain unclear. Our study investigates a novel acetylation/deacetylation switch in TAZ, governing its subcellular localization in melanoma tumorigenesis. METHODS Immunoprecipitation/Western blot assessed TAZ protein interactions and acetylation. SIRT5 activity was quantified with enzyme-linked immunosorbent assay. Immunofluorescence indicated TAZ nuclear localization. TEAD transcriptional activity was measured through luciferase reporter assays. ChIP detected TAZ binding to the CTGF promoter. Transwell and wound healing assays quantified melanoma cell invasiveness and migration. Metastasis was evaluated using a mouse model via tail vein injections. Clinical relevance was explored via immunohistochemical staining of patient tumors. RESULTS CBP facilitated TAZ acetylation at K54 in response to epidermal growth factor stimulation, while SIRT5 mediated deacetylation. Acetylation correlated with phosphorylation, regulating TAZ's binding with LATS2 or TEAD. TAZ K54 acetylation enhanced its S89 phosphorylation, promoting cytosolic retention via LATS2 interaction. SIRT5-mediated deacetylation enhanced TAZ-TEAD interaction and nuclear retention. Chromatin IP showed SIRT5-deacetylated TAZ recruited to CTGF promoter, boosting transcriptional activity. In a mouse model, SIRT5 overexpression induced melanoma metastasis to lung tissue following the injection of B16F10 melanocytes via the tail vein, and this effect was prevented by verteporfin treatment. CONCLUSIONS Our study revealed a novel mechanism of TAZ nuclear retention regulated by SIRT5-mediated K54 deacetylation and demonstrated the significance of TAZ deacetylation in CTGF expression. This study highlights the potential implications of the SIRT5/TAZ axis for treating metastatic melanoma.
Collapse
Affiliation(s)
- Garam Kim
- College of Pharmacy, Chosun University, 309 Pilmun-daero, Dong-gu, Gwangju, 501-759, Republic of Korea
| | - Poshan Yugal Bhattarai
- College of Pharmacy, Chosun University, 309 Pilmun-daero, Dong-gu, Gwangju, 501-759, Republic of Korea
| | - Sung-Chul Lim
- Department of Pathology, School of Medicine, Chosun University, Gwangju, 61452, Republic of Korea
| | - Kwang Youl Lee
- College of Pharmacy, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Hong Seok Choi
- College of Pharmacy, Chosun University, 309 Pilmun-daero, Dong-gu, Gwangju, 501-759, Republic of Korea.
| |
Collapse
|
28
|
Azumah R, Hummitzsch K, Anderson RA, Rodgers RJ. Expression of transforming growth factor β signalling molecules and their correlations with genes in loci linked to polycystic ovary syndrome in human foetal and adult tissues. Reprod Fertil Dev 2024; 36:RD23174. [PMID: 38894494 DOI: 10.1071/rd23174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 05/20/2024] [Indexed: 06/21/2024] Open
Abstract
Context Altered signalling of androgens, anti-Müllerian hormone or transforming growth factor beta (TGFβ) during foetal development have been implicated in the predisposition to polycystic ovary syndrome (PCOS) in later life, aside from its genetic predisposition. In foetal ovarian fibroblasts, TGFβ1 has been shown to regulate androgen signalling and seven genes located in loci associated with PCOS. Since PCOS exhibits a myriad of symptoms, it likely involves many different organs. Aims To identify the relationships between TGFβ signalling molecules and PCOS candidate genes in different tissues associated with PCOS. Methods Using RNA sequencing data, we examined the expression patterns of TGFβ signalling molecules in the human ovary, testis, heart, liver, kidney, brain tissue, and cerebellum from 4 to 20weeks of gestation and postnatally. We also examined the correlations between gene expression of TGFβ signalling molecules and PCOS candidate genes. Key results TGFβ signalling molecules were dynamically expressed in most tissues prenatally and/or postnatally. FBN3 , a PCOS candidate gene involved in TGFβ signalling, was expressed during foetal development in all tissues. The PCOS candidate genes HMGA2, YAP1 , and RAD50 correlated significantly (P TGFBR1 in six out of the seven tissues examined. Conclusions This study suggests that possible crosstalk occurs between genes in loci associated with PCOS and TGFβ signalling molecules in multiple tissues, particularly during foetal development. Implications Thus, alteration in TGFβ signalling during foetal development could affect many tissues contributing to the multiple phenotypes of PCOS in later life.
Collapse
Affiliation(s)
- Rafiatu Azumah
- Robinson Research Institute, School of Biomedicine, The University of Adelaide, Adelaide, SA 5005, Australia
| | - Katja Hummitzsch
- Robinson Research Institute, School of Biomedicine, The University of Adelaide, Adelaide, SA 5005, Australia
| | - Richard A Anderson
- Medical Research Council Centre for Reproductive Health, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Raymond J Rodgers
- Robinson Research Institute, School of Biomedicine, The University of Adelaide, Adelaide, SA 5005, Australia
| |
Collapse
|
29
|
Zhang Y, Ren Y, Li X, Li M, Fu M, Zhou W, Yu Y, Xiong Y. A review on decoding the roles of YAP/TAZ signaling pathway in cardiovascular diseases: Bridging molecular mechanisms to therapeutic insights. Int J Biol Macromol 2024; 271:132473. [PMID: 38795886 DOI: 10.1016/j.ijbiomac.2024.132473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 05/02/2024] [Accepted: 05/15/2024] [Indexed: 05/28/2024]
Abstract
Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) serve as transcriptional co-activators that dynamically shuttle between the cytoplasm and nucleus, resulting in either the suppression or enhancement of their downstream gene expression. Recent emerging evidence demonstrates that YAP/TAZ is strongly implicated in the pathophysiological processes that contribute to cardiovascular diseases (CVDs). In the cardiovascular system, YAP/TAZ is involved in the orchestration of a range of biological processes such as oxidative stress, inflammation, proliferation, and autophagy. Furthermore, YAP/TAZ has been revealed to be closely associated with the initiation and development of various cardiovascular diseases, including atherosclerosis, pulmonary hypertension, myocardial fibrosis, cardiac hypertrophy, and cardiomyopathy. In this review, we delve into recent studies surrounding YAP and TAZ, along with delineating their roles in contributing to the pathogenesis of CVDs with a link to various physiological processes in the cardiovascular system. Additionally, we highlight the current potential drugs targeting YAP/TAZ for CVDs therapy and discuss their challenges for translational application. Overall, this review may offer novel insights for understanding and treating cardiovascular disorders.
Collapse
Affiliation(s)
- Yan Zhang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an 710069, Shaanxi, PR China
| | - Yuanyuan Ren
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an 710069, Shaanxi, PR China
| | - Xiaofang Li
- Department of Gastroenterology, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Xi'an, Shaanxi 710018, PR China
| | - Man Li
- Department of Endocrinology, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Xi'an, Shaanxi 710018, PR China
| | - Mingdi Fu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an 710069, Shaanxi, PR China
| | - Wenjing Zhou
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an 710069, Shaanxi, PR China
| | - Yi Yu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an 710069, Shaanxi, PR China.
| | - Yuyan Xiong
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an 710069, Shaanxi, PR China; Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, the Affiliated Hospital of Northwest University, 710018 Xi'an, Shaanxi, PR China.
| |
Collapse
|
30
|
Hemba-Waduge RUS, Liu M, Li X, Sun JL, Budslick EA, Bondos SE, Ji JY. Metabolic control by the Bithorax Complex-Wnt signaling crosstalk in Drosophila. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.31.596851. [PMID: 38853890 PMCID: PMC11160800 DOI: 10.1101/2024.05.31.596851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Adipocytes distributed throughout the body play crucial roles in lipid metabolism and energy homeostasis. Regional differences among adipocytes influence normal function and disease susceptibility, but the mechanisms driving this regional heterogeneity remain poorly understood. Here, we report a genetic crosstalk between the Bithorax Complex ( BX-C ) genes and Wnt/Wingless signaling that orchestrates regional differences among adipocytes in Drosophila larvae. Abdominal adipocytes, characterized by the exclusive expression of abdominal A ( abd-A ) and Abdominal B ( Abd-B ), exhibit distinct features compared to thoracic adipocytes, with Wnt signaling further amplifying these disparities. Depletion of BX-C genes in adipocytes reduces fat accumulation, delays larval-pupal transition, and eventually leads to pupal lethality. Depleting Abd-A or Abd-B reduces Wnt target gene expression, thereby attenuating Wnt signaling-induced lipid mobilization. Conversely, Wnt signaling stimulated abd-A transcription, suggesting a feedforward loop that amplifies the interplay between Wnt signaling and BX-C in adipocytes. These findings elucidate how the crosstalk between cell-autonomous BX-C gene expression and Wnt signaling define unique metabolic behaviors in adipocytes in different anatomical regions of fat body, delineating larval adipose tissue domains.
Collapse
|
31
|
Liang H, Xu Y, Zhao J, Chen M, Wang M. Hippo pathway in non-small cell lung cancer: mechanisms, potential targets, and biomarkers. Cancer Gene Ther 2024; 31:652-666. [PMID: 38499647 PMCID: PMC11101353 DOI: 10.1038/s41417-024-00761-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 03/04/2024] [Accepted: 03/07/2024] [Indexed: 03/20/2024]
Abstract
Lung cancer is the primary contributor to cancer-related deaths globally, and non-small cell lung cancer (NSCLC) constitutes around 85% of all lung cancer cases. Recently, the emergence of targeted therapy and immunotherapy revolutionized the treatment of NSCLC and greatly improved patients' survival. However, drug resistance is inevitable, and extensive research has demonstrated that the Hippo pathway plays a crucial role in the development of drug resistance in NSCLC. The Hippo pathway is a highly conserved signaling pathway that is essential for various biological processes, including organ development, maintenance of epithelial balance, tissue regeneration, wound healing, and immune regulation. This pathway exerts its effects through two key transcription factors, namely Yes-associated protein (YAP) and transcriptional co-activator PDZ-binding motif (TAZ). They regulate gene expression by interacting with the transcriptional-enhanced associate domain (TEAD) family. In recent years, this pathway has been extensively studied in NSCLC. The review summarizes a comprehensive overview of the involvement of this pathway in NSCLC, and discusses the mechanisms of drug resistance, potential targets, and biomarkers associated with this pathway in NSCLC.
Collapse
Affiliation(s)
- Hongge Liang
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yan Xu
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jing Zhao
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Minjiang Chen
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Mengzhao Wang
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
32
|
Biswal P, Sahu MR, Ahmad MH, Mondal AC. The interplay between hippo signaling and mitochondrial metabolism: Implications for cellular homeostasis and disease. Mitochondrion 2024; 76:101885. [PMID: 38643865 DOI: 10.1016/j.mito.2024.101885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 04/10/2024] [Accepted: 04/18/2024] [Indexed: 04/23/2024]
Abstract
Mitochondria are the membrane-bound organelles producing energy for cellular metabolic processes. They orchestrate diverse cell signaling cascades regulating cellular homeostasis. This functional versatility may be attributed to their ability to regulate mitochondrial dynamics, biogenesis, and apoptosis. The Hippo pathway, a conserved signaling pathway, regulates various cellular processes, including mitochondrial functions. Through its effectors YAP and TAZ, the Hippo pathway regulates transcription factors and creates a seriatim process that mediates cellular metabolism, mitochondrial dynamics, and survival. Mitochondrial dynamics also potentially regulates Hippo signaling activation, indicating a bidirectional relationship between the two. This review outlines the interplay between the Hippo signaling components and the multifaceted role of mitochondria in cellular homeostasis under physiological and pathological conditions.
Collapse
Affiliation(s)
- Priyanka Biswal
- Laboratory of Cellular and Molecular Neurobiology, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Manas Ranjan Sahu
- Laboratory of Cellular and Molecular Neurobiology, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Mir Hilal Ahmad
- Laboratory of Cellular and Molecular Neurobiology, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Amal Chandra Mondal
- Laboratory of Cellular and Molecular Neurobiology, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India.
| |
Collapse
|
33
|
Jung O, Baek MJ, Wooldrik C, Johnson KR, Fisher KW, Lou J, Ricks TJ, Wen T, Best MD, Cryns VL, Anderson RA, Choi S. Nuclear phosphoinositide signaling promotes YAP/TAZ-TEAD transcriptional activity in breast cancer. EMBO J 2024; 43:1740-1769. [PMID: 38565949 PMCID: PMC11066040 DOI: 10.1038/s44318-024-00085-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 02/29/2024] [Accepted: 03/08/2024] [Indexed: 04/04/2024] Open
Abstract
The Hippo pathway effectors Yes-associated protein 1 (YAP) and its homolog TAZ are transcriptional coactivators that control gene expression by binding to TEA domain (TEAD) family transcription factors. The YAP/TAZ-TEAD complex is a key regulator of cancer-specific transcriptional programs, which promote tumor progression in diverse types of cancer, including breast cancer. Despite intensive efforts, the YAP/TAZ-TEAD complex in cancer has remained largely undruggable due to an incomplete mechanistic understanding. Here, we report that nuclear phosphoinositides function as cofactors that mediate the binding of YAP/TAZ to TEADs. The enzymatic products of phosphoinositide kinases PIPKIα and IPMK, including phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2) and phosphatidylinositol 3,4,5-trisphosphate (P(I3,4,5)P3), bridge the binding of YAP/TAZ to TEAD. Inhibiting these kinases or the association of YAP/TAZ with PI(4,5)P2 and PI(3,4,5)P3 attenuates YAP/TAZ interaction with the TEADs, the expression of YAP/TAZ target genes, and breast cancer cell motility. Although we could not conclusively exclude the possibility that other enzymatic products of IPMK such as inositol phosphates play a role in the mechanism, our results point to a previously unrecognized role of nuclear phosphoinositide signaling in control of YAP/TAZ activity and implicate this pathway as a potential therapeutic target in YAP/TAZ-driven breast cancer.
Collapse
Affiliation(s)
- Oisun Jung
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Min-Jeong Baek
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
- Interdisciplinary Graduate Program in Biomedical Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Colin Wooldrik
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
- Interdisciplinary Graduate Program in Biomedical Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Keith R Johnson
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
- Department of Oral Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Kurt W Fisher
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Jinchao Lou
- Department of Chemistry, University of Tennessee, 1420 Circle Drive, Knoxville, TN, 37996, USA
| | - Tanei J Ricks
- Department of Chemistry, University of Memphis, 3744 Walker Avenue, Memphis, TN, 38152, USA
| | - Tianmu Wen
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Michael D Best
- Department of Chemistry, University of Tennessee, 1420 Circle Drive, Knoxville, TN, 37996, USA
| | - Vincent L Cryns
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Richard A Anderson
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Suyong Choi
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA.
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
34
|
Kashihara T, Sadoshima J. Regulation of myocardial glucose metabolism by YAP/TAZ signaling. J Cardiol 2024; 83:323-329. [PMID: 38266816 DOI: 10.1016/j.jjcc.2024.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Accepted: 01/12/2024] [Indexed: 01/26/2024]
Abstract
The heart utilizes glucose and its metabolites as both energy sources and building blocks for cardiac growth and survival under both physiological and pathophysiological conditions. YAP/TAZ, transcriptional co-activators of the Hippo pathway, are key regulators of cell proliferation, survival, and metabolism in many cell types. Increasing lines of evidence suggest that the Hippo-YAP/TAZ signaling pathway is involved in the regulation of both physiological and pathophysiological processes in the heart. In particular, YAP/TAZ play a critical role in mediating aerobic glycolysis, the Warburg effect, in cardiomyocytes. Here, we summarize what is currently known about YAP/TAZ signaling in the heart by focusing on the regulation of glucose metabolism and its functional significance.
Collapse
Affiliation(s)
- Toshihide Kashihara
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, Tokyo, Japan
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ, USA.
| |
Collapse
|
35
|
Miskin RP, DiPersio CM. Roles for epithelial integrin α3β1 in regulation of the microenvironment during normal and pathological tissue remodeling. Am J Physiol Cell Physiol 2024; 326:C1308-C1319. [PMID: 38497112 PMCID: PMC11371326 DOI: 10.1152/ajpcell.00128.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 03/08/2024] [Accepted: 03/08/2024] [Indexed: 03/19/2024]
Abstract
Integrin receptors for the extracellular matrix activate intracellular signaling pathways that are critical for tissue development, homeostasis, and regeneration/repair, and their loss or dysregulation contributes to many developmental defects and tissue pathologies. This review will focus on tissue remodeling roles for integrin α3β1, a receptor for laminins found in the basement membranes (BMs) that underlie epithelial cell layers. As a paradigm, we will discuss literature that supports a role for α3β1 in promoting ability of epidermal keratinocytes to modify their tissue microenvironment during skin development, wound healing, or tumorigenesis. Preclinical and clinical studies have shown that this role depends largely on ability of α3β1 to govern the keratinocyte's repertoire of secreted proteins, or the "secretome," including 1) matrix proteins and proteases involved in matrix remodeling and 2) paracrine-acting growth factors/cytokines that stimulate other cells with important tissue remodeling functions (e.g., endothelial cells, fibroblasts, inflammatory cells). Moreover, α3β1 signaling controls gene expression that helps epithelial cells carry out these functions, including genes that encode secreted matrix proteins, proteases, growth factors, or cytokines. We will review what is known about α3β1-dependent gene regulation through both transcription and posttranscriptional mRNA stability. Regarding the latter, we will discuss examples of α3β1-dependent alternative splicing (AS) or alternative polyadenylation (APA) that prevents inclusion of cis-acting mRNA sequences that would otherwise target the transcript for degradation via nonsense-mediated decay or destabilizing AU-rich elements (AREs) in the 3'-untranslated region (3'-UTR). Finally, we will discuss prospects and anticipated challenges of exploiting α3β1 as a clinical target for the treatment of cancer or wound healing.
Collapse
Affiliation(s)
| | - C Michael DiPersio
- Department of Surgery, Albany Medical College, Albany, New York, United States
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York, United States
| |
Collapse
|
36
|
Pan C, Hao X, Deng X, Lu F, Liu J, Hou W, Xu T. The roles of Hippo/YAP signaling pathway in physical therapy. Cell Death Discov 2024; 10:197. [PMID: 38670949 PMCID: PMC11053014 DOI: 10.1038/s41420-024-01972-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 04/16/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
Cellular behavior is regulated by mechanical signals within the cellular microenvironment. Additionally, changes of temperature, blood flow, and muscle contraction also affect cellular state and the development of diseases. In clinical practice, physical therapy techniques such as ultrasound, vibration, exercise, cold therapy, and hyperthermia are commonly employed to alleviate pain and treat diseases. However, the molecular mechanism about how these physiotherapy methods stimulate local tissues and control gene expression remains unknow. Fortunately, the discovery of YAP filled this gap, which has been reported has the ability to sense and convert a wide variety of mechanical signals into cell-specific programs for transcription, thereby offering a fresh perspective on the mechanisms by which physiotherapy treat different diseases. This review examines the involvement of Hippo/YAP signaling pathway in various diseases and its role in different physical therapy approaches on diseases. Furthermore, we explore the potential therapeutic implications of the Hippo/YAP signaling pathway and address the limitations and controversies surrounding its application in physiotherapy.
Collapse
Affiliation(s)
- Chunran Pan
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoxia Hao
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaofeng Deng
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fan Lu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiawei Liu
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenjie Hou
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tao Xu
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
37
|
Pfeifer M, Brammeld JS, Price S, Pilling J, Bhavsar D, Farcas A, Bateson J, Sundarrajan A, Miragaia RJ, Guan N, Arnold S, Tariq L, Grondine M, Talbot S, Guerriero ML, O'Neill DJ, Young J, Company C, Dunn S, Thorpe H, Martin MJ, Maratea K, Barrell D, Ahdesmaki M, Mettetal JT, Brownell J, McDermott U. Genome-wide CRISPR screens identify the YAP/TEAD axis as a driver of persister cells in EGFR mutant lung cancer. Commun Biol 2024; 7:497. [PMID: 38658677 PMCID: PMC11043391 DOI: 10.1038/s42003-024-06190-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 04/12/2024] [Indexed: 04/26/2024] Open
Abstract
Most lung cancer patients with metastatic cancer eventually relapse with drug-resistant disease following treatment and EGFR mutant lung cancer is no exception. Genome-wide CRISPR screens, to either knock out or overexpress all protein-coding genes in cancer cell lines, revealed the landscape of pathways that cause resistance to the EGFR inhibitors osimertinib or gefitinib in EGFR mutant lung cancer. Among the most recurrent resistance genes were those that regulate the Hippo pathway. Following osimertinib treatment a subpopulation of cancer cells are able to survive and over time develop stable resistance. These 'persister' cells can exploit non-genetic (transcriptional) programs that enable cancer cells to survive drug treatment. Using genetic and pharmacologic tools we identified Hippo signalling as an important non-genetic mechanism of cell survival following osimertinib treatment. Further, we show that combinatorial targeting of the Hippo pathway and EGFR is highly effective in EGFR mutant lung cancer cells and patient-derived organoids, suggesting a new therapeutic strategy for EGFR mutant lung cancer patients.
Collapse
Affiliation(s)
- Matthias Pfeifer
- Oncology R&D, AstraZeneca, 1 Francis Crick Avenue, Cambridge, CB2 0RE, UK
- Leibniz-Institute of Virology (LIV) and University hospital Hamburg-Eppendorf (UKE), Hamburg, Germany
| | | | - Stacey Price
- Wellcome Sanger Institute, Hinxton, Cambridge, CB10 1SA, UK
| | - James Pilling
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, 1 Francis Crick Avenue, Cambridge, CB2 0RE, UK
| | - Deepa Bhavsar
- Oncology R&D, AstraZeneca, 1 Francis Crick Avenue, Cambridge, CB2 0RE, UK
| | - Anca Farcas
- Oncology R&D, AstraZeneca, 1 Francis Crick Avenue, Cambridge, CB2 0RE, UK
| | | | - Anjana Sundarrajan
- Oncology R&D, AstraZeneca, 1 Francis Crick Avenue, Cambridge, CB2 0RE, UK
| | - Ricardo J Miragaia
- Oncology R&D, AstraZeneca, 1 Francis Crick Avenue, Cambridge, CB2 0RE, UK
| | - Nin Guan
- Oncology R&D, AstraZeneca, 1 Francis Crick Avenue, Cambridge, CB2 0RE, UK
| | - Stephanie Arnold
- Oncology R&D, AstraZeneca, 1 Francis Crick Avenue, Cambridge, CB2 0RE, UK
| | - Laiba Tariq
- Oncology R&D, AstraZeneca, 1 Francis Crick Avenue, Cambridge, CB2 0RE, UK
| | - Michael Grondine
- Oncology R&D, AstraZeneca, 1 Francis Crick Avenue, Cambridge, CB2 0RE, UK
| | - Sarah Talbot
- Oncology R&D, AstraZeneca, 1 Francis Crick Avenue, Cambridge, CB2 0RE, UK
| | - Maria Lisa Guerriero
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, 1 Francis Crick Avenue, Cambridge, CB2 0RE, UK
| | - Daniel J O'Neill
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, 1 Francis Crick Avenue, Cambridge, CB2 0RE, UK
| | - Jamie Young
- Wellcome Sanger Institute, Hinxton, Cambridge, CB10 1SA, UK
| | - Carlos Company
- Oncology R&D, AstraZeneca, 1 Francis Crick Avenue, Cambridge, CB2 0RE, UK
| | - Shanade Dunn
- Oncology R&D, AstraZeneca, 1 Francis Crick Avenue, Cambridge, CB2 0RE, UK
| | - Hannah Thorpe
- Oncology R&D, AstraZeneca, 1 Francis Crick Avenue, Cambridge, CB2 0RE, UK
| | - Matthew J Martin
- Oncology R&D, AstraZeneca, 1 Francis Crick Avenue, Cambridge, CB2 0RE, UK
| | - Kimberly Maratea
- Clinical Pharmacology & Safety, BioPharmaceuticals R&D, AstraZeneca, 1 Francis Crick Avenue, Cambridge, CB2 0RE, UK
| | - Daniel Barrell
- Oncology R&D, AstraZeneca, 1 Francis Crick Avenue, Cambridge, CB2 0RE, UK
| | - Miika Ahdesmaki
- Oncology R&D, AstraZeneca, 1 Francis Crick Avenue, Cambridge, CB2 0RE, UK
| | - Jerome T Mettetal
- Oncology R&D, AstraZeneca, 1 Francis Crick Avenue, Cambridge, CB2 0RE, UK
| | - James Brownell
- Oncology R&D, AstraZeneca, 1 Francis Crick Avenue, Cambridge, CB2 0RE, UK
| | - Ultan McDermott
- Oncology R&D, AstraZeneca, 1 Francis Crick Avenue, Cambridge, CB2 0RE, UK.
| |
Collapse
|
38
|
Song M, Wang H, Liu C, Jin S, Liu B, Sun W. Non-coding RNAs as regulators of the Hippo pathway in cardiac development and cardiovascular disease. Front Pharmacol 2024; 15:1348280. [PMID: 38698813 PMCID: PMC11063341 DOI: 10.3389/fphar.2024.1348280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 04/09/2024] [Indexed: 05/05/2024] Open
Abstract
Cardiovascular diseases pose a serious threat to human health. The onset of cardiovascular diseases involves the comprehensive effects of multiple genes and environmental factors, and multiple signaling pathways are involved in regulating the occurrence and development of cardiovascular diseases. The Hippo pathway is a highly conserved signaling pathway involved in the regulation of cell proliferation, apoptosis, and differentiation. Recently, it has been widely studied in the fields of cardiovascular disease, cancer, and cell regeneration. Non-coding RNA (ncRNAs), which are important small molecules for the regulation of gene expression in cells, can directly target genes and have diverse regulatory functions. Recent studies have found that ncRNAs interact with Hippo pathway components to regulate myocardial fibrosis, cardiomyocyte proliferation, apoptosis, and hypertrophy and play an important role in cardiovascular disease. In this review, we describe the mode of action of ncRNAs in regulating the Hippo pathway, provide new ideas for further research, and identify molecules involved in the mechanism of action of ncRNAs and the Hippo pathway as potential therapeutic targets, with the aim of finding new modes of action for the treatment and prevention of cardiovascular diseases.
Collapse
Affiliation(s)
- Mengyang Song
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, China
| | - He Wang
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, China
| | - Caixia Liu
- Department of Neurology, The Liaoning Province People’s Hospital, Shenyang, China
| | - Sijie Jin
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, China
| | - Bin Liu
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, China
| | - Wei Sun
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
39
|
So CL, Robitaille M, Sadras F, McCullough MH, Milevskiy MJG, Goodhill GJ, Roberts-Thomson SJ, Monteith GR. Cellular geometry and epithelial-mesenchymal plasticity intersect with PIEZO1 in breast cancer cells. Commun Biol 2024; 7:467. [PMID: 38632473 PMCID: PMC11024093 DOI: 10.1038/s42003-024-06163-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Accepted: 04/08/2024] [Indexed: 04/19/2024] Open
Abstract
Differences in shape can be a distinguishing feature between different cell types, but the shape of a cell can also be dynamic. Changes in cell shape are critical when cancer cells escape from the primary tumor and undergo major morphological changes that allow them to squeeze between endothelial cells, enter the vasculature, and metastasize to other areas of the body. A shift from rounded to spindly cellular geometry is a consequence of epithelial-mesenchymal plasticity, which is also associated with changes in gene expression, increased invasiveness, and therapeutic resistance. However, the consequences and functional impacts of cell shape changes and the mechanisms through which they occur are still poorly understood. Here, we demonstrate that altering the morphology of a cell produces a remodeling of calcium influx via the ion channel PIEZO1 and identify PIEZO1 as an inducer of features of epithelial-to-mesenchymal plasticity. Combining automated epifluorescence microscopy and a genetically encoded calcium indicator, we demonstrate that activation of the PIEZO1 force channel with the PIEZO1 agonist, YODA 1, induces features of epithelial-to-mesenchymal plasticity in breast cancer cells. These findings suggest that PIEZO1 is a critical point of convergence between shape-induced changes in cellular signaling and epithelial-mesenchymal plasticity in breast cancer cells.
Collapse
Affiliation(s)
- Choon Leng So
- School of Pharmacy, The University of Queensland, Woolloongabba, QLD, 4102, Australia
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Mélanie Robitaille
- School of Pharmacy, The University of Queensland, Woolloongabba, QLD, 4102, Australia
| | - Francisco Sadras
- School of Pharmacy, The University of Queensland, Woolloongabba, QLD, 4102, Australia
| | - Michael H McCullough
- Queensland Brain Institute and School of Mathematics and Physics, The University of Queensland, Brisbane, QLD, 4072, Australia
- Eccles Institute of Neuroscience, John Curtin School of Medical Research, and School of Computing, ANU College of Engineering and Computer Science, The Australian National University, Canberra, ACT, 2600, Australia
| | - Michael J G Milevskiy
- ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 2010, Australia
| | - Geoffrey J Goodhill
- Queensland Brain Institute and School of Mathematics and Physics, The University of Queensland, Brisbane, QLD, 4072, Australia
- Departments of Developmental Biology and Neuroscience, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | | | - Gregory R Monteith
- School of Pharmacy, The University of Queensland, Woolloongabba, QLD, 4102, Australia.
| |
Collapse
|
40
|
Bruno T, Catena V, Corleone G, Cortile C, Cappelletto MC, Bellei B, De Nicola F, Amadio B, Gumenyuk S, Marchesi F, Annibali O, Blandino G, Fanciulli M, Di Agostino S. Che-1/miR-590-3p/TAZ axis sustains multiple myeloma disease. Leukemia 2024; 38:877-882. [PMID: 38368441 PMCID: PMC10997508 DOI: 10.1038/s41375-024-02168-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 02/01/2024] [Accepted: 02/02/2024] [Indexed: 02/19/2024]
Affiliation(s)
- Tiziana Bruno
- SAFU Laboratory, Department of Research, Advanced Diagnostics, and Technological Innovation, Translational Research Area, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Valeria Catena
- SAFU Laboratory, Department of Research, Advanced Diagnostics, and Technological Innovation, Translational Research Area, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Giacomo Corleone
- SAFU Laboratory, Department of Research, Advanced Diagnostics, and Technological Innovation, Translational Research Area, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Clelia Cortile
- SAFU Laboratory, Department of Research, Advanced Diagnostics, and Technological Innovation, Translational Research Area, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | | | - Barbara Bellei
- Laboratory of Cutaneous Physiopathology and Integrated Center of Metabolomics Research, IRCCS San Gallicano Dermatological Institute, Rome, Italy
| | - Francesca De Nicola
- SAFU Laboratory, Department of Research, Advanced Diagnostics, and Technological Innovation, Translational Research Area, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Bruno Amadio
- SAFU Laboratory, Department of Research, Advanced Diagnostics, and Technological Innovation, Translational Research Area, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Svitlana Gumenyuk
- Hematology and Stem Cell Transplant Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Francesco Marchesi
- Hematology and Stem Cell Transplant Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Ombretta Annibali
- Unit Of Hematology, Stem Cell Transplantation, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128, Roma, Italy
| | - Giovanni Blandino
- Translational Oncology Research Unit, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi, 53, 00144, Rome, Italy
| | - Maurizio Fanciulli
- SAFU Laboratory, Department of Research, Advanced Diagnostics, and Technological Innovation, Translational Research Area, IRCCS Regina Elena National Cancer Institute, Rome, Italy.
| | - Silvia Di Agostino
- Department of Health Sciences, Magna Græcia University of Catanzaro, 88100, Catanzaro, Italy
| |
Collapse
|
41
|
Melrose J. Hippo cell signaling and HS-proteoglycans regulate tissue form and function, age-dependent maturation, extracellular matrix remodeling, and repair. Am J Physiol Cell Physiol 2024; 326:C810-C828. [PMID: 38223931 DOI: 10.1152/ajpcell.00683.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/09/2024] [Accepted: 01/09/2024] [Indexed: 01/16/2024]
Abstract
This review examined how Hippo cell signaling and heparan sulfate (HS)-proteoglycans (HSPGs) regulate tissue form and function. Despite being a nonweight-bearing tissue, the brain is regulated by Hippo mechanoresponsive cell signaling pathways during embryonic development. HS-proteoglycans interact with growth factors, morphogens, and extracellular matrix components to regulate development and pathology. Pikachurin and Eyes shut (Eys) interact with dystroglycan to stabilize the photoreceptor axoneme primary cilium and ribbon synapse facilitating phototransduction and neurotransduction with bipolar retinal neuronal networks in ocular vision, the primary human sense. Another HSPG, Neurexin interacts with structural and adaptor proteins to stabilize synapses and ensure specificity of neural interactions, and aids in synaptic potentiation and plasticity in neurotransduction. HSPGs also stabilize the blood-brain barrier and motor neuron basal structures in the neuromuscular junction. Agrin and perlecan localize acetylcholinesterase and its receptors in the neuromuscular junction essential for neuromuscular control. The primary cilium is a mechanosensory hub on neurons, utilized by YES associated protein (YAP)-transcriptional coactivator with PDZ-binding motif (TAZ) Hippo, Hh, Wnt, transforming growth factor (TGF)-β/bone matrix protein (BMP) receptor tyrosine kinase cell signaling. Members of the glypican HSPG proteoglycan family interact with Smoothened and Patched G-protein coupled receptors on the cilium to regulate Hh and Wnt signaling during neuronal development. Control of glycosyl sulfotransferases and endogenous protease expression by Hippo TAZ YAP represents a mechanism whereby the fine structure of HS-proteoglycans can be potentially modulated spatiotemporally to regulate tissue morphogenesis in a similar manner to how Hippo signaling controls sialyltransferase expression and mediation of cell-cell recognition, dysfunctional sialic acid expression is a feature of many tumors.
Collapse
Affiliation(s)
- James Melrose
- Raymond Purves Laboratory, Institute of Bone and Joint Research, Kolling Institute of Medical Research, University of Sydney, Northern Sydney Local Health District, Royal North Shore Hospital, St. Leonards, New South Wales, Australia
- Sydney Medical School-Northern, University of Sydney at Royal North Shore Hospital, St. Leonards, New South Wales, Australia
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
42
|
Longmate WM, Norton E, Duarte GA, Wu L, DiPersio MR, Lamar JM, DiPersio CM. Keratinocyte integrin α3β1 induces expression of the macrophage stimulating factor, CSF-1, through a YAP/TEAD-dependent mechanism. Matrix Biol 2024; 127:48-56. [PMID: 38340968 PMCID: PMC10923166 DOI: 10.1016/j.matbio.2024.02.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 01/11/2024] [Accepted: 02/05/2024] [Indexed: 02/12/2024]
Abstract
The development of wound therapy targeting integrins is hampered by inadequate understanding of integrin function in cutaneous wound healing and the wound microenvironment. Following cutaneous injury, keratinocytes migrate to restore the skin barrier, and macrophages aid in debris clearance. Thus, both keratinocytes and macrophages are critical to the coordination of tissue repair. Keratinocyte integrins have been shown to participate in this coordinated effort by regulating secreted factors, some of which crosstalk to distinct cells in the wound microenvironment. Epidermal integrin α3β1 is a receptor for laminin-332 in the cutaneous basement membrane. Here we show that wounds deficient in epidermal α3β1 express less epidermal-derived macrophage colony-stimulating factor 1 (CSF-1), the primary macrophage-stimulating growth factor. α3β1-deficient wounds also have fewer wound-proximal macrophages, suggesting that keratinocyte α3β1 may stimulate wound macrophages through the regulation of CSF-1. Indeed, using a set of immortalized keratinocytes, we demonstrate that keratinocyte-derived CSF-1 supports macrophage growth, and that α3β1 regulates Csf1 expression through Src-dependent stimulation of Yes-associated protein (YAP)-Transcriptional enhanced associate domain (TEAD)-mediated transcription. Consistently, α3β1-deficient wounds in vivo display a substantially reduced number of keratinocytes with YAP-positive nuclei. Overall, our current findings identify a novel role for epidermal integrin α3β1 in regulating the cutaneous wound microenvironment by mediating paracrine crosstalk from keratinocytes to wound macrophages, implicating α3β1 as a potential target of wound therapy.
Collapse
Affiliation(s)
- Whitney M Longmate
- Department of Surgery, Albany Medical College, Albany, NY 12208, USA; Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA
| | - Emily Norton
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA
| | - Giesse Albeche Duarte
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA
| | - Lei Wu
- Department of Surgery, Albany Medical College, Albany, NY 12208, USA
| | - Mathieu R DiPersio
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA
| | - John M Lamar
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA
| | - C Michael DiPersio
- Department of Surgery, Albany Medical College, Albany, NY 12208, USA; Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY 12208, USA.
| |
Collapse
|
43
|
Sadri F, Hosseini SF, Rezaei Z, Fereidouni M. Hippo-YAP/TAZ signaling in breast cancer: Reciprocal regulation of microRNAs and implications in precision medicine. Genes Dis 2024; 11:760-771. [PMID: 37692482 PMCID: PMC10491881 DOI: 10.1016/j.gendis.2023.01.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 12/09/2022] [Accepted: 01/29/2023] [Indexed: 09/12/2023] Open
Abstract
Breast cancer is a molecularly heterogeneous disease and the most common female malignancy. In recent years, therapy approaches have evolved to accommodate molecular diversity, with a focus on more biologically based therapies to minimize negative consequences. To regulate cell fate in human breast cells, the Hippo signaling pathway has been associated with the alpha subtype of estrogen receptors. This pathway regulates tissue size, regeneration, and healing, as well as the survival of tissue-specific stem cells, proliferation, and apoptosis in a variety of organs, allowing for cell differentiation. Hippo signaling is mediated by the kinases MST1, MST2, LATS1, and LATS2, as well as the adaptor proteins SAV1 and MOB. These kinases phosphorylate the downstream effectors of the Hippo pathway, yes-associated protein (YAP), and transcriptional coactivator with PDZ-binding motif (TAZ), suppressing the expression of their downstream target genes. The Hippo signaling pathway kinase cascade plays a significant role in all cancers. Understanding the principles of this kinase cascade would prevent the occurrence of breast cancer. In recent years, small noncoding RNAs, or microRNAs, have been implicated in the development of several malignancies, including breast cancer. The interconnections between miRNAs and Hippo signaling pathway core proteins in the breast, on the other hand, remain poorly understood. In this review, we focused on highlighting the Hippo signaling system, its key parts, its importance in breast cancer, and its regulation by miRNAs and other related pathways.
Collapse
Affiliation(s)
- Farzad Sadri
- Student Research Committee, Birjand University of Medical Sciences, Birjand 9717853577, Iran
| | | | - Zohreh Rezaei
- Department of Biology, University of Sistan and Baluchestan, Zahedan 9816745785, Iran
| | - Mohammad Fereidouni
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand 9717853577, Iran
| |
Collapse
|
44
|
Crossey E, Carty S, Shao F, Henao-Vasquez J, Ysasi AB, Zeng M, Hinds A, Lo M, Tilston-Lunel A, Varelas X, Jones MR, Fine A. Influenza Induces Lung Lymphangiogenesis Independent of YAP/TAZ Activity in Lymphatic Endothelial Cells. RESEARCH SQUARE 2024:rs.3.rs-3951689. [PMID: 38463972 PMCID: PMC10925403 DOI: 10.21203/rs.3.rs-3951689/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
The lymphatic system consists of a vessel network lined by specialized lymphatic endothelial cells (LECs) that are responsible for tissue fluid homeostasis and immune cell trafficking. The mechanisms for organ-specific LEC responses to environmental cues are not well understood. We found robust lymphangiogenesis during influenza A virus infection in the adult mouse lung. We show that the number of LECs increases 2-fold at 7 days post-influenza infection (dpi) and 3-fold at 21 dpi, and that lymphangiogenesis is preceded by lymphatic dilation. We also show that the expanded lymphatic network enhances fluid drainage to mediastinal lymph nodes. Using EdU labeling, we found that a significantly higher number of pulmonary LECs are proliferating at 7 dpi compared to LECs in homeostatic conditions. Lineage tracing during influenza indicates that new pulmonary LECs are derived from preexisting LECs rather than non-LEC progenitors. Lastly, using a conditional LEC-specific YAP/TAZ knockout model, we established that lymphangiogenesis, fluid transport and the immune response to influenza are independent of YAP/TAZ activity in LECs. These findings were unexpected, as they indicate that YAP/TAZ signaling is not crucial for these processes.
Collapse
Affiliation(s)
- Erin Crossey
- Boston University Chobanian and Avedisian School of Medicine
| | - Senegal Carty
- Boston University Chobanian and Avedisian School of Medicine
| | - Fengzhi Shao
- Boston University Chobanian and Avedisian School of Medicine
| | | | | | - Michelle Zeng
- Boston University Chobanian and Avedisian School of Medicine
| | - Anne Hinds
- Boston University Chobanian and Avedisian School of Medicine
| | - Ming Lo
- Boston University Chobanian and Avedisian School of Medicine
| | | | | | - Matthew R Jones
- Boston University Chobanian and Avedisian School of Medicine
| | - Alan Fine
- Boston University Chobanian and Avedisian School of Medicine
| |
Collapse
|
45
|
Wu J, Qin C, Tian F, Liu X, Hu J, Wu F, Chen C, Lin Y. Epigenetic drug screening for trophoblast syncytialization reveals a novel role for MLL1 in regulating fetoplacental growth. BMC Med 2024; 22:57. [PMID: 38317232 PMCID: PMC10845764 DOI: 10.1186/s12916-024-03264-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Accepted: 01/18/2024] [Indexed: 02/07/2024] Open
Abstract
BACKGROUND Abnormal placental development is a significant factor contributing to perinatal morbidity and mortality, affecting approximately 5-7% of pregnant women. Trophoblast syncytialization plays a pivotal role in the establishment and maturation of the placenta, and its dysregulation is closely associated with several pregnancy-related disorders, including preeclampsia and intrauterine growth restriction. However, the underlying mechanisms and genetic determinants of syncytialization are largely unknown. METHODS We conducted a systematic drug screen using an epigenetic compound library to systematically investigate the epigenetic mechanism essential for syncytialization, and identified mixed lineage leukemia 1 (MLL1), a histone 3 lysine 4 methyltransferase, as a crucial regulator of trophoblast syncytialization. BeWo cells were utilized to investigate the role of MLL1 during trophoblast syncytialization. RNA sequencing and CUT&Tag were further performed to search for potential target genes and the molecular pathways involved. Human placenta tissue was used to investigate the role of MLL1 in TEA domain transcription factor 4 (TEAD4) expression and the upstream signaling during syncytialization. A mouse model was used to examine whether inhibition of MLL1-mediated H3K4me3 regulated placental TEAD4 expression and fetoplacental growth. RESULTS Genetic knockdown of MLL1 or pharmacological inhibition of the MLL1 methyltransferase complex (by MI-3454) markedly enhanced syncytialization, while overexpression of MLL1 inhibited forskolin (FSK)-induced syncytiotrophoblast formation. In human placental villous tissue, MLL1 was predominantly localized in the nuclei of cytotrophoblasts. Moreover, a notable upregulation in MLL1 expression was observed in the villus tissue of patients with preeclampsia compared with that in the control group. Based on RNA sequencing and CUT&Tag analyses, depletion of MLL1 inhibited the Hippo signaling pathway by suppressing TEAD4 expression by modulating H3K4me3 levels on the TEAD4 promoter region. TEAD4 overexpression significantly reversed the FSK-induced or MLL1 silencing-mediated trophoblast syncytialization. Additionally, decreased hypoxia-inducible factor 1A (HIF1A) enrichment at the MLL1 promoter was observed during syncytialization. Under hypoxic conditions, HIF1A could bind to and upregulate MLL1, leading to the activation of the MLL1/TEAD4 axis. In vivo studies demonstrated that the administration of MI-3454 significantly enhanced fetal vessel development and increased the thickness of the syncytial layer, thereby supporting fetoplacental growth. CONCLUSIONS These results revealed a novel epigenetic mechanism underlying the progression of syncytialization with MLL1, and suggest potential avenues for identifying new therapeutic targets for pregnancy-related disorders.
Collapse
Affiliation(s)
- Jiayi Wu
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
- Institute of Birth Defects and Rare Diseases, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Chuanmei Qin
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
- Institute of Birth Defects and Rare Diseases, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Fuju Tian
- Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xueqing Liu
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
- Institute of Birth Defects and Rare Diseases, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jianing Hu
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
- Institute of Birth Defects and Rare Diseases, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Fan Wu
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
- Institute of Birth Defects and Rare Diseases, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Cailian Chen
- Department of Automation, Shanghai Jiao Tong University, Key Laboratory of System Control and Information Processing, Ministry of Education of China, Shanghai, China
| | - Yi Lin
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.
| |
Collapse
|
46
|
Rajagopalan K, Christyraj JDS, Chelladurai KS, Das P, Mahendran K, Nagarajan L, Gunalan S. Understanding the Multi-Functional Role of TCTP in the Regeneration Process of Earthworm, Perionyx excavatus. Tissue Eng Regen Med 2024; 21:353-366. [PMID: 37935935 PMCID: PMC10825100 DOI: 10.1007/s13770-023-00599-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 08/30/2023] [Accepted: 09/18/2023] [Indexed: 11/09/2023] Open
Abstract
BACKGROUND Regeneration is a highly complex process that requires the coordination of numerous molecular events, and identifying the key ruler that governs is important to investigate. While it has been shown that TCTP is a multi-functional protein that regulates cell proliferation, differentiation, apoptosis, anti-apoptosis, stem cell maintenance, and immune responses, but only a few studies associated to regeneration have been reported. To investigate the multi-functional role of TCTP in regeneration, the earthworm Perionyx excavatus was chosen. METHODS Through pharmacological suppression of TCTP, amputation, histology, molecular docking, and western blotting, the multi-function role of TCTP involved in regeneration is revealed. RESULTS Amputational studies show that P. excavatus is a clitellum-independent regenerating earthworm resulting in two functional worms upon amputation. Arresting cell cycle at the G1/S boundary using 2 mM Thymidine confirms that P. excavatus execute both epimorphosis and morphallaxis regeneration mode. The pharmacological suppression of TCTP using buclizine results in regeneration suppression. Following the combinatorial injection of 2 mM Thymidine and buclizine, the earthworm regeneration is completely blocked, which suggests a critical functional role of TCTP in morphallaxis. The pharmacological inhibition of TCTP also suppresses the key proteins involved in regeneration: Wnt3a (stem cell marker), PCNA (cell proliferation) and YAP1 (Hippo signalling) but augments the expression of cellular stress protein p53. CONCLUSION The collective results indicate that TCTP synchronously is involved in the process of stem cell activation, cell proliferation, morphallaxis, and organ development in the regeneration event.
Collapse
Affiliation(s)
- Kamarajan Rajagopalan
- Molecular Biology and Stem Cell Research Lab, Centre for Molecular and Nanomedical Sciences, International Research Centre, Sathyabama Institute of Science and Technology (Deemed to be University), Chennai, Tamilnadu, India
| | - Jackson Durairaj Selvan Christyraj
- Molecular Biology and Stem Cell Research Lab, Centre for Molecular and Nanomedical Sciences, International Research Centre, Sathyabama Institute of Science and Technology (Deemed to be University), Chennai, Tamilnadu, India.
| | - Karthikeyan Subbiahanadar Chelladurai
- Molecular Biology and Stem Cell Research Lab, Centre for Molecular and Nanomedical Sciences, International Research Centre, Sathyabama Institute of Science and Technology (Deemed to be University), Chennai, Tamilnadu, India
- School of Health Sciences, Purdue University, 550 Stadium Mall Drive, West Lafayette, IN, 47907, USA
| | - Puja Das
- Molecular Biology and Stem Cell Research Lab, Centre for Molecular and Nanomedical Sciences, International Research Centre, Sathyabama Institute of Science and Technology (Deemed to be University), Chennai, Tamilnadu, India
| | - Karthikeyan Mahendran
- Department of Zoology and Microbiology, Thiyagarajar College, Madurai, Tamilnadu, India
| | - Logeshwari Nagarajan
- Molecular Biology and Stem Cell Research Lab, Centre for Molecular and Nanomedical Sciences, International Research Centre, Sathyabama Institute of Science and Technology (Deemed to be University), Chennai, Tamilnadu, India
| | - Saritha Gunalan
- Molecular Biology and Stem Cell Research Lab, Centre for Molecular and Nanomedical Sciences, International Research Centre, Sathyabama Institute of Science and Technology (Deemed to be University), Chennai, Tamilnadu, India
| |
Collapse
|
47
|
Huang F, Wei G, Wang H, Zhang Y, Lan W, Xie Y, Wu G. Fibroblasts inhibit osteogenesis by regulating nuclear-cytoplasmic shuttling of YAP in mesenchymal stem cells and secreting DKK1. Biol Res 2024; 57:4. [PMID: 38245803 PMCID: PMC10799393 DOI: 10.1186/s40659-023-00481-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Accepted: 12/04/2023] [Indexed: 01/22/2024] Open
Abstract
BACKGROUND Fibrous scars frequently form at the sites of bone nonunion when attempts to repair bone fractures have failed. However, the detailed mechanism by which fibroblasts, which are the main components of fibrous scars, impede osteogenesis remains largely unknown. RESULTS In this study, we found that fibroblasts compete with osteogenesis in both human bone nonunion tissues and BMP2-induced ectopic osteogenesis in a mouse model. Fibroblasts could inhibit the osteoblastic differentiation of mesenchymal stem cells (MSCs) via direct and indirect cell competition. During this process, fibroblasts modulated the nuclear-cytoplasmic shuttling of YAP in MSCs. Knocking down YAP could inhibit osteoblast differentiation of MSCs, while overexpression of nuclear-localized YAP-5SA could reverse the inhibition of osteoblast differentiation of MSCs caused by fibroblasts. Furthermore, fibroblasts secreted DKK1, which further inhibited the formation of calcium nodules during the late stage of osteogenesis but did not affect the early stage of osteogenesis. Thus, fibroblasts could inhibit osteogenesis by regulating YAP localization in MSCs and secreting DKK1. CONCLUSIONS Our research revealed that fibroblasts could modulate the nuclear-cytoplasmic shuttling of YAP in MSCs, thereby inhibiting their osteoblast differentiation. Fibroblasts could also secrete DKK1, which inhibited calcium nodule formation at the late stage of osteogenesis.
Collapse
Affiliation(s)
- Fei Huang
- Central Laboratory, First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, Fujian, China
| | - Guozhen Wei
- Department of Orthopaedics, The First Affiliated Hospital, Fujian Medical University, No. 20, Chazhong Road, Taijiang District, Fuzhou, 350005, Fujian, China
- Department of Orthopaedics, Binhai Campus of the First Affiliated Hospital, National Regional Medical Center, Fujian Medical University, Fuzhou, 350212, Fujian, China
| | - Hai Wang
- Department of Orthopaedics, The First Affiliated Hospital, Fujian Medical University, No. 20, Chazhong Road, Taijiang District, Fuzhou, 350005, Fujian, China
- Department of Orthopaedics, Binhai Campus of the First Affiliated Hospital, National Regional Medical Center, Fujian Medical University, Fuzhou, 350212, Fujian, China
| | - Ying Zhang
- Central Laboratory, First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, Fujian, China
| | - Wenbin Lan
- Department of Orthopaedics, The First Affiliated Hospital, Fujian Medical University, No. 20, Chazhong Road, Taijiang District, Fuzhou, 350005, Fujian, China
- Department of Orthopaedics, Binhai Campus of the First Affiliated Hospital, National Regional Medical Center, Fujian Medical University, Fuzhou, 350212, Fujian, China
| | - Yun Xie
- Department of Orthopaedics, The First Affiliated Hospital, Fujian Medical University, No. 20, Chazhong Road, Taijiang District, Fuzhou, 350005, Fujian, China.
- Department of Orthopaedics, Binhai Campus of the First Affiliated Hospital, National Regional Medical Center, Fujian Medical University, Fuzhou, 350212, Fujian, China.
| | - Gui Wu
- Department of Orthopaedics, The First Affiliated Hospital, Fujian Medical University, No. 20, Chazhong Road, Taijiang District, Fuzhou, 350005, Fujian, China.
- Department of Orthopaedics, Binhai Campus of the First Affiliated Hospital, National Regional Medical Center, Fujian Medical University, Fuzhou, 350212, Fujian, China.
| |
Collapse
|
48
|
Hillen H, Candi A, Vanderhoydonck B, Kowalczyk W, Sansores-Garcia L, Kesikiadou EC, Van Huffel L, Spiessens L, Nijs M, Soons E, Haeck W, Klaassen H, Smets W, Spieser SA, Marchand A, Chaltin P, Ciesielski F, Debaene F, Chen L, Kamal A, Gwaltney SL, Versele M, Halder GA. A Novel Irreversible TEAD Inhibitor, SWTX-143, Blocks Hippo Pathway Transcriptional Output and Causes Tumor Regression in Preclinical Mesothelioma Models. Mol Cancer Ther 2024; 23:3-13. [PMID: 37748190 DOI: 10.1158/1535-7163.mct-22-0681] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 03/13/2023] [Accepted: 08/30/2023] [Indexed: 09/27/2023]
Abstract
The Hippo pathway and its downstream effectors, the YAP and TAZ transcriptional coactivators, are deregulated in multiple different types of human cancer and are required for cancer cell phenotypes in vitro and in vivo, while largely dispensable for tissue homeostasis in adult mice. YAP/TAZ and their main partner transcription factors, the TEAD1-4 factors, are therefore promising anticancer targets. Because of frequent YAP/TAZ hyperactivation caused by mutations in the Hippo pathway components NF2 and LATS2, mesothelioma is one of the prime cancer types predicted to be responsive to YAP/TAZ-TEAD inhibitor treatment. Mesothelioma is a devastating disease for which currently no effective treatment options exist. Here, we describe a novel covalent YAP/TAZ-TEAD inhibitor, SWTX-143, that binds to the palmitoylation pocket of all four TEAD isoforms. SWTX-143 caused irreversible and specific inhibition of the transcriptional activity of YAP/TAZ-TEAD in Hippo-mutant tumor cell lines. More importantly, YAP/TAZ-TEAD inhibitor treatment caused strong mesothelioma regression in subcutaneous xenograft models with human cells and in an orthotopic mesothelioma mouse model. Finally, SWTX-143 also selectively impaired the growth of NF2-mutant kidney cancer cell lines, suggesting that the sensitivity of mesothelioma models to these YAP/TAZ-TEAD inhibitors can be extended to other tumor types with aberrations in Hippo signaling. In brief, we describe a novel and specific YAP/TAZ-TEAD inhibitor that has potential to treat multiple Hippo-mutant solid tumor types.
Collapse
Affiliation(s)
- Hanne Hillen
- VIB Center for Cancer Biology and KU Leuven Department of Oncology, KU Leuven, Leuven, Belgium
| | | | | | - Weronika Kowalczyk
- VIB Center for Cancer Biology and KU Leuven Department of Oncology, KU Leuven, Leuven, Belgium
| | - Leticia Sansores-Garcia
- VIB Center for Cancer Biology and KU Leuven Department of Oncology, KU Leuven, Leuven, Belgium
| | - Elena C Kesikiadou
- VIB Center for Cancer Biology and KU Leuven Department of Oncology, KU Leuven, Leuven, Belgium
| | - Leen Van Huffel
- VIB Center for Cancer Biology and KU Leuven Department of Oncology, KU Leuven, Leuven, Belgium
| | - Lore Spiessens
- VIB Center for Cancer Biology and KU Leuven Department of Oncology, KU Leuven, Leuven, Belgium
| | | | | | | | | | | | | | | | - Patrick Chaltin
- Cistim Leuven vzw, Leuven, Belgium
- Center for Drug Design and Discovery (CD3), KU Leuven, Leuven, Belgium
| | | | | | - Lei Chen
- SpringWorks Therapeutics, Stamford, Connecticut
| | | | | | | | - Georg A Halder
- VIB Center for Cancer Biology and KU Leuven Department of Oncology, KU Leuven, Leuven, Belgium
| |
Collapse
|
49
|
Payne S, Neal A, De Val S. Transcription factors regulating vasculogenesis and angiogenesis. Dev Dyn 2024; 253:28-58. [PMID: 36795082 PMCID: PMC10952167 DOI: 10.1002/dvdy.575] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 02/06/2023] [Accepted: 02/06/2023] [Indexed: 02/17/2023] Open
Abstract
Transcription factors (TFs) play a crucial role in regulating the dynamic and precise patterns of gene expression required for the initial specification of endothelial cells (ECs), and during endothelial growth and differentiation. While sharing many core features, ECs can be highly heterogeneous. Differential gene expression between ECs is essential to pattern the hierarchical vascular network into arteries, veins and capillaries, to drive angiogenic growth of new vessels, and to direct specialization in response to local signals. Unlike many other cell types, ECs have no single master regulator, instead relying on differing combinations of a necessarily limited repertoire of TFs to achieve tight spatial and temporal activation and repression of gene expression. Here, we will discuss the cohort of TFs known to be involved in directing gene expression during different stages of mammalian vasculogenesis and angiogenesis, with a primary focus on development.
Collapse
Affiliation(s)
- Sophie Payne
- Department of Physiology, Anatomy and GeneticsInstitute of Developmental and Regenerative Medicine, University of OxfordOxfordUK
| | - Alice Neal
- Department of Physiology, Anatomy and GeneticsInstitute of Developmental and Regenerative Medicine, University of OxfordOxfordUK
| | - Sarah De Val
- Department of Physiology, Anatomy and GeneticsInstitute of Developmental and Regenerative Medicine, University of OxfordOxfordUK
| |
Collapse
|
50
|
Zhao X, Li D, Song Y, Xu J, Xiang FL. Drug Discovery for Adult Cardiomyocyte Regeneration: Opportunities and Challenges. Antioxid Redox Signal 2023; 39:1070-1087. [PMID: 37166381 DOI: 10.1089/ars.2023.0319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Significance: Cardiovascular disease is a major contributor to human mortality and morbidity. The cardiac tissue undergoes fibrotic healing after injury because of the limited regenerative capacity of adult mammalian cardiomyocyte (CM). Extensive research has been performed to identify therapeutic targets for CM regeneration, as the success of promoting adult human CM regeneration to repair the injured heart is considered the Holy Grail in the field. Recent Advances: To date, more than 30 target genes have been shown to regulate adult mammalian CM proliferation. More than 20 targets have been validated in adult mouse myocardial infarction (MI) model in a therapeutic setting. In this review, the translational efficacy readouts from 17 selected pharmaceutical targets are summarized, among which the Hippo-yes-associated protein (Yap) pathway is the most extensively investigated and fits the criteria for a promising target for pro-CM-regeneration therapy development. Critical Issues and Future Directions: As the pro-CM-regeneration potential of current drug treatment for cardiovascular patients is limited, to help identify and fill the gap between basic research and drug discovery in this specific field, details regarding target identification, validation in mouse MI models, high-throughput screening assay development, and preclinical in vivo efficacy model optimization are discussed. Finally, suggestions and recommendations are also provided to help establish a common guideline for in vivo translational studies for drug discovery focusing on CM regeneration. Antioxid. Redox Signal. 39, 1070-1087.
Collapse
Affiliation(s)
- Xu Zhao
- Department of Anesthesiology and the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Donghua Li
- Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yiyan Song
- Department of Anesthesiology and the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jie Xu
- Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Fu-Li Xiang
- Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|