1
|
Lim WY, Lee JH, Choi Y, Yoon K. Verteporfin is an effective inhibitor of HCMV replication. Virus Res 2024; 350:199475. [PMID: 39362410 PMCID: PMC11492081 DOI: 10.1016/j.virusres.2024.199475] [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: 06/16/2024] [Revised: 09/20/2024] [Accepted: 09/26/2024] [Indexed: 10/05/2024]
Abstract
Human cytomegalovirus (HCMV), a double-stranded DNA virus from the Betaherpesvirinae subfamily, constitutes significant risks to newborns and immunocompromised individuals, potentially leading to severe neurodevelopmental disorders. The purpose of this study was to identify FDA-approved drugs that can inhibit HCMV replication through a drug repositioning approach. Using an HCMV progeny assay, verteporfin, a medication used as a photosensitizer in photodynamic therapy, was found to inhibit HCMV production in a dose-dependent manner, significantly reducing replication at concentrations as low as 0.5 µM, approximately 1/20th of the concentration used in anti-cancer research. Further analysis revealed that verteporfin did not interfere with HCMV host cell entry or nuclear transport but reduced viral mRNA and protein levels throughout the HCMV life cycle from the immediate-early stages. These results suggest that verteporfin has the potential to be rapidly and safely developed as a repurposed drug to inhibit HCMV infection.
Collapse
Affiliation(s)
- Woo Young Lim
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon, Gyeonggi-do 16419, South Korea
| | - Ju Hyun Lee
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon, Gyeonggi-do 16419, South Korea
| | - Youngju Choi
- Department of Microbiology, School of Medicine, Sungkyunkwan University, Suwon, Gyeonggi-do 16419, South Korea
| | - Keejung Yoon
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon, Gyeonggi-do 16419, South Korea; College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, Gyeonggi-do 16419, South Korea.
| |
Collapse
|
2
|
Wang XW, Yang ZY, Li T, Zhao XR, Li XZ, Wang XX. Verteporfin Exerts Anticancer Effects and Reverses Resistance to Paclitaxel via Inducing Ferroptosis in Esophageal Squamous Cell Cancer Cells. Mol Biotechnol 2024; 66:2558-2568. [PMID: 37751128 DOI: 10.1007/s12033-023-00891-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: 03/01/2023] [Accepted: 09/06/2023] [Indexed: 09/27/2023]
Abstract
Esophageal squamous cell carcinoma (ESCC) is one of the most common malignant tumors. Ferroptosis is a new form of regulated cell death and targeting ferroptosis provides a novel therapeutic approach for human cancers. Verteporfin (VP) has been identified as a Yes-associated protein (YAP) inhibitor for treatment of several human cancers. However, it remains unclear whether VP exerts anticancer activity by inducing ferroptosis in ESCC cells. In the current study, we found that VP reduced cell viability and led to cell death in ESCC cell lines (KYSE150 and KYSE30) by inhibiting YAP expression. Subsequently, the findings revealed that VP treatment triggered significant ferroptosis events, including accumulation of Fe2+, reactive oxygen species (ROS) and malondialdehyde (MDA), reduction of mitochondrial membrane potential (MMP), glutathione (GSH) and glutathione peroxidase 4 (GPX4) expression. Further study showed that the effects of ESCC cell proliferation and death caused by VP could be reversed by ferroptosis inhibitor ferrostatin-1 (Fer-1). Moreover, VP enhanced the chemosensitivity of ESCC resistant cells to paclitaxel (PTX). And VP combined with PTX can synergistically inhibit cell proliferation and induce cell death by triggering ferroptosis of PTX-resistant cells. All these data suggested that VP suppressed ESCC cell survival and reversed resistance to PTX through inducing ferroptosis, which may provide a promising therapeutic strategy for ESCC.
Collapse
Affiliation(s)
- Xue-Wei Wang
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, 030001, China
| | - Zi-Yi Yang
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, 030001, China
| | - Ting Li
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, 030001, China
| | - Xin-Ran Zhao
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, 030001, China
| | - Xiao-Zhong Li
- Department of Infectious Diseases, Shanxi Provincial People's Hospital, Affiliated People's Hospital of Shanxi Medical University, Taiyuan, 030012, China.
| | - Xiao-Xia Wang
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, 030001, China.
| |
Collapse
|
3
|
Wang XX, Zhao YM, Zhang QY, Zhao JX, Yin DH, Zhang ZZ, Jin XY, Li SN, Ji HY, Chen HY, Guo XF, Yu Y, Ma WY, Yan H, Li H, Ou-Yang QM, Pan ZW, Liang HH, Wang N, Chen W, Cai BZ, Liu Y. Acetylcytidine modification of Amotl1 by N-acetyltransferase 10 contributes to cardiac fibrotic expansion in mice after myocardial infarction. Acta Pharmacol Sin 2024; 45:1425-1437. [PMID: 38839936 PMCID: PMC11192918 DOI: 10.1038/s41401-024-01306-8] [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: 01/08/2024] [Accepted: 05/06/2024] [Indexed: 06/07/2024] Open
Abstract
Cardiac fibrosis is a pathological scarring process that impairs cardiac function. N-acetyltransferase 10 (Nat10) is recently identified as the key enzyme for the N4-acetylcytidine (ac4C) modification of mRNAs. In this study, we investigated the role of Nat10 in cardiac fibrosis following myocardial infarction (MI) and the related mechanisms. MI was induced in mice by ligation of the left anterior descending coronary artery; cardiac function was assessed with echocardiography. We showed that both the mRNA and protein expression levels of Nat10 were significantly increased in the infarct zone and border zone 4 weeks post-MI, and the expression of Nat10 in cardiac fibroblasts was significantly higher compared with that in cardiomyocytes after MI. Fibroblast-specific overexpression of Nat10 promoted collagen deposition and induced cardiac systolic dysfunction post-MI in mice. Conversely, fibroblast-specific knockout of Nat10 markedly relieved cardiac function impairment and extracellular matrix remodeling following MI. We then conducted ac4C-RNA binding protein immunoprecipitation-sequencing (RIP-seq) in cardiac fibroblasts transfected with Nat10 siRNA, and revealed that angiomotin-like 1 (Amotl1), an upstream regulator of the Hippo signaling pathway, was the target gene of Nat10. We demonstrated that Nat10-mediated ac4C modification of Amotl1 increased its mRNA stability and translation in neonatal cardiac fibroblasts, thereby increasing the interaction of Amotl1 with yes-associated protein 1 (Yap) and facilitating Yap translocation into the nucleus. Intriguingly, silencing of Amotl1 or Yap, as well as treatment with verteporfin, a selective and potent Yap inhibitor, attenuated the Nat10 overexpression-induced proliferation of cardiac fibroblasts and prevented their differentiation into myofibroblasts in vitro. In conclusion, this study highlights Nat10 as a crucial regulator of myocardial fibrosis following MI injury through ac4C modification of upstream activators within the Hippo/Yap signaling pathway.
Collapse
Affiliation(s)
- Xiu-Xiu Wang
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology at College of Pharmacy (National Key Laboratory of Frigid Zone Cardiovascular Diseases, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, 150086, China
- Institute of Clinical Pharmacology (The Heilongjiang Key Laboratory of Drug Research), Harbin Medical University, Harbin, 150086, China
| | - Yi-Ming Zhao
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology at College of Pharmacy (National Key Laboratory of Frigid Zone Cardiovascular Diseases, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, 150086, China
- Institute of Clinical Pharmacology (The Heilongjiang Key Laboratory of Drug Research), Harbin Medical University, Harbin, 150086, China
| | - Qian-Yun Zhang
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology at College of Pharmacy (National Key Laboratory of Frigid Zone Cardiovascular Diseases, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, 150086, China
- Institute of Clinical Pharmacology (The Heilongjiang Key Laboratory of Drug Research), Harbin Medical University, Harbin, 150086, China
| | - Jing-Xuan Zhao
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology at College of Pharmacy (National Key Laboratory of Frigid Zone Cardiovascular Diseases, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, 150086, China
- Institute of Clinical Pharmacology (The Heilongjiang Key Laboratory of Drug Research), Harbin Medical University, Harbin, 150086, China
| | - Dao-Hong Yin
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology at College of Pharmacy (National Key Laboratory of Frigid Zone Cardiovascular Diseases, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, 150086, China
- Institute of Clinical Pharmacology (The Heilongjiang Key Laboratory of Drug Research), Harbin Medical University, Harbin, 150086, China
| | - Zi-Zhen Zhang
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology at College of Pharmacy (National Key Laboratory of Frigid Zone Cardiovascular Diseases, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, 150086, China
- Institute of Clinical Pharmacology (The Heilongjiang Key Laboratory of Drug Research), Harbin Medical University, Harbin, 150086, China
| | - Xiao-Yan Jin
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology at College of Pharmacy (National Key Laboratory of Frigid Zone Cardiovascular Diseases, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, 150086, China
| | - Shuai-Nan Li
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology at College of Pharmacy (National Key Laboratory of Frigid Zone Cardiovascular Diseases, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, 150086, China
| | - Hao-Yu Ji
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology at College of Pharmacy (National Key Laboratory of Frigid Zone Cardiovascular Diseases, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, 150086, China
- Institute of Clinical Pharmacology (The Heilongjiang Key Laboratory of Drug Research), Harbin Medical University, Harbin, 150086, China
| | - Hong-Yang Chen
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology at College of Pharmacy (National Key Laboratory of Frigid Zone Cardiovascular Diseases, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, 150086, China
- Institute of Clinical Pharmacology (The Heilongjiang Key Laboratory of Drug Research), Harbin Medical University, Harbin, 150086, China
| | - Xiao-Fei Guo
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology at College of Pharmacy (National Key Laboratory of Frigid Zone Cardiovascular Diseases, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, 150086, China
- Institute of Clinical Pharmacology (The Heilongjiang Key Laboratory of Drug Research), Harbin Medical University, Harbin, 150086, China
| | - Yang Yu
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology at College of Pharmacy (National Key Laboratory of Frigid Zone Cardiovascular Diseases, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, 150086, China
- Institute of Clinical Pharmacology (The Heilongjiang Key Laboratory of Drug Research), Harbin Medical University, Harbin, 150086, China
| | - Wen-Ya Ma
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology at College of Pharmacy (National Key Laboratory of Frigid Zone Cardiovascular Diseases, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, 150086, China
- Institute of Clinical Pharmacology (The Heilongjiang Key Laboratory of Drug Research), Harbin Medical University, Harbin, 150086, China
| | - Hong Yan
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology at College of Pharmacy (National Key Laboratory of Frigid Zone Cardiovascular Diseases, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, 150086, China
| | - Han Li
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology at College of Pharmacy (National Key Laboratory of Frigid Zone Cardiovascular Diseases, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, 150086, China
- Institute of Clinical Pharmacology (The Heilongjiang Key Laboratory of Drug Research), Harbin Medical University, Harbin, 150086, China
| | - Qi-Meng Ou-Yang
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology at College of Pharmacy (National Key Laboratory of Frigid Zone Cardiovascular Diseases, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, 150086, China
- Institute of Clinical Pharmacology (The Heilongjiang Key Laboratory of Drug Research), Harbin Medical University, Harbin, 150086, China
| | - Zhen-Wei Pan
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology at College of Pharmacy (National Key Laboratory of Frigid Zone Cardiovascular Diseases, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, 150086, China
| | - Hai-Hai Liang
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology at College of Pharmacy (National Key Laboratory of Frigid Zone Cardiovascular Diseases, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, 150086, China
| | - Ning Wang
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology at College of Pharmacy (National Key Laboratory of Frigid Zone Cardiovascular Diseases, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, 150086, China.
| | - Wei Chen
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China.
| | - Ben-Zhi Cai
- Department of Pharmacy at the Second Affiliated Hospital, and Department of Pharmacology at College of Pharmacy (National Key Laboratory of Frigid Zone Cardiovascular Diseases, Key Laboratory of Cardiovascular Research, Ministry of Education), Harbin Medical University, Harbin, 150086, China.
- Institute of Clinical Pharmacology (The Heilongjiang Key Laboratory of Drug Research), Harbin Medical University, Harbin, 150086, China.
- NHC Key Laboratory of Cell Transplantation, The Heilongjiang Key Laboratory of Drug Research, Harbin Medical University, Harbin, 150001, China.
| | - Yu Liu
- Department of Clinical Laboratory at the Fourth Affiliated Hospital of Harbin Medical University, Harbin, 150001, China.
| |
Collapse
|
4
|
Fu J, Liu W, Liu S, Zhao R, Hayashi T, Zhao H, Xiang Y, Mizuno K, Hattori S, Fujisaki H, Ikejima T. Inhibition of YAP/TAZ pathway contributes to the cytotoxicity of silibinin in MCF-7 and MDA-MB-231 human breast cancer cells. Cell Signal 2024; 119:111186. [PMID: 38643945 DOI: 10.1016/j.cellsig.2024.111186] [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: 01/29/2024] [Revised: 04/14/2024] [Accepted: 04/18/2024] [Indexed: 04/23/2024]
Abstract
Breast cancer is one of the most common cancers threatening women's health. Our previous study found that silibinin induced the death of MCF-7 and MDA-MB-231 human breast cancer cells. We noticed that silibinin-induced cell damage was accompanied by morphological changes, including the increased cell aspect ratio (cell length/width) and decreased cell area. Besides, the cytoskeleton is also destroyed in cells treated with silibinin. YAP/TAZ, a mechanical signal sensor interacted with extracellular pressure, cell adhesion area and cytoskeleton, is also closely associated with cell survival, proliferation and migration. Thus, the involvement of YAP/TAZ in the cytotoxicity of silibinin in breast cancer cells has attracted our interests. Excitingly, we find that silibinin inhibits the nuclear translocation of YAP/TAZ in MCF-7 and MDA-MB-231 cells, and reduces the mRNA expressions of YAP/TAZ target genes, ACVR1, MnSOD and ANKRD. More importantly, expression of YAP1 gene is negatively correlated with the survival of the patients with breast cancers. Molecular docking analysis reveals high probabilities for binding of silibinin to the proteins in the YAP pathways. DARTS and CETSA results confirm the binding abilities of silibinin to YAP and LATS. Inhibiting YAP pathway either by addition of verteporfin, an inhibitor of YAP/TAZ-TEAD, or by transfection of si-RNAs targeting YAP or TAZ further enhances silibinin-induced cell damage. While enhancing YAP activity by silencing LATS1/2 or overexpressing YAPS127/397A, an active form of YAP, attenuates silibinin-induced cell damage. These findings demonstrate that inhibition of the YAP/TAZ pathway contributes to cytotoxicity of silibinin in breast cancers, shedding lights on YAP/TAZ-targeted cancer therapies.
Collapse
Affiliation(s)
- Jianing Fu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Weiwei Liu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Siyu Liu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Ruxiao Zhao
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Toshihiko Hayashi
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China; Nippi Research Institute of Biomatrix, Toride, Ibaraki 302-0017, Japan
| | - Haina Zhao
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Yinlanqi Xiang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Kazunori Mizuno
- Nippi Research Institute of Biomatrix, Toride, Ibaraki 302-0017, Japan
| | - Shunji Hattori
- Nippi Research Institute of Biomatrix, Toride, Ibaraki 302-0017, Japan
| | - Hitomi Fujisaki
- Nippi Research Institute of Biomatrix, Toride, Ibaraki 302-0017, Japan
| | - Takashi Ikejima
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China; Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning, China.
| |
Collapse
|
5
|
Nong J, Shen S, Hong F, Xiao F, Meng L, Li P, Lei X, Chen YG. Verteporfin inhibits TGF-β signaling by disrupting the Smad2/3-Smad4 interaction. Mol Biol Cell 2024; 35:ar95. [PMID: 38696259 PMCID: PMC11244160 DOI: 10.1091/mbc.e24-02-0073] [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: 02/20/2024] [Revised: 04/15/2024] [Accepted: 04/22/2024] [Indexed: 05/04/2024] Open
Abstract
Transforming growth factor-β (TGF-β) signaling plays a crucial role in pathogenesis, such as accelerating tissue fibrosis and promoting tumor development at the later stages of tumorigenesis by promoting epithelial-mesenchymal transition (EMT), cancer cell migration, and invasion. Targeting TGF-β signaling is a promising therapeutic approach, but nonspecific inhibition may result in adverse effects. In this study, we focus on the Smad2/3-Smad4 complex, a key component in TGF-β signaling transduction, as a potential target for cancer therapy. Through a phase-separated condensate-aided biomolecular interaction system, we identified verteporfin (VP) as a small-molecule inhibitor that specifically targets the Smad2/3-Smad4 interaction. VP effectively disrupted the interaction between Smad2/3 and Smad4 and thereby inhibited canonical TGF-β signaling, but not the interaction between Smad1 and Smad4 in bone morphogenetic protein (BMP) signaling. Furthermore, VP exhibited inhibitory effects on TGF-β-induced EMT and cell migration. Our findings indicate a novel approach to develop protein-protein interaction inhibitors of the canonical TGF-β signaling pathway for treatments of related diseases.
Collapse
Affiliation(s)
- Junxiu Nong
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Shengqiang Shen
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Department of Chemical Biology, College of Chemistry and Molecular Engineering, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Fan Hong
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangzhou 510005, Guangdong Province, China
| | - Fan Xiao
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Department of Chemical Biology, College of Chemistry and Molecular Engineering, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Lingtian Meng
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Pilong Li
- Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Xiaoguang Lei
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Department of Chemical Biology, College of Chemistry and Molecular Engineering, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Ye-Guang Chen
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangzhou 510005, Guangdong Province, China
- School of Basic Medicine, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
| |
Collapse
|
6
|
Hao S, Lee YJ, Benhamou Goldfajn N, Flores E, Liang J, Fuehrer H, Demmerle J, Lippincott-Schwartz J, Liu Z, Sukenik S, Cai D. YAP condensates are highly organized hubs. iScience 2024; 27:109927. [PMID: 38784009 PMCID: PMC11111833 DOI: 10.1016/j.isci.2024.109927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 10/24/2023] [Accepted: 05/03/2024] [Indexed: 05/25/2024] Open
Abstract
YAP/TEAD signaling is essential for organismal development, cell proliferation, and cancer progression. As a transcriptional coactivator, how YAP activates its downstream target genes is incompletely understood. YAP forms biomolecular condensates in response to hyperosmotic stress, concentrating transcription-related factors to activate downstream target genes. However, whether YAP forms condensates under other signals, how YAP condensates organize and function, and how YAP condensates activate transcription in general are unknown. Here, we report that endogenous YAP forms sub-micron scale condensates in response to Hippo pathway regulation and actin cytoskeletal tension. YAP condensates are stabilized by the transcription factor TEAD1, and recruit BRD4, a coactivator that is enriched at active enhancers. Using single-particle tracking, we found that YAP condensates slowed YAP diffusion within condensate boundaries, a possible mechanism for promoting YAP target search. These results reveal that YAP condensate formation is a highly regulated process that is critical for YAP/TEAD target gene expression.
Collapse
Affiliation(s)
- Siyuan Hao
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Ye Jin Lee
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Nadav Benhamou Goldfajn
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
- Department of Biophysics, Johns Hopkins University, 3400 N. Charles St., Baltimore, MD 21218, USA
| | - Eduardo Flores
- Department of Chemistry and Chemical Biology, University of California, Merced, Merced, CA 95343, USA
| | - Jindayi Liang
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Hannah Fuehrer
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Justin Demmerle
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | | | - Zhe Liu
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Shahar Sukenik
- Department of Chemistry and Chemical Biology, University of California, Merced, Merced, CA 95343, USA
| | - Danfeng Cai
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| |
Collapse
|
7
|
Cao Z, Hou Y, Zhao Z, Zhang H, Tian L, Zhang Y, Dong C, Guo F, Tan L, Han Y, Wang W, Jiao S, Tang Y, An L, Zhou Z. Reactivating Hippo by drug compounds to suppress gastric cancer and enhance chemotherapy sensitivity. J Biol Chem 2024; 300:107311. [PMID: 38657866 PMCID: PMC11126936 DOI: 10.1016/j.jbc.2024.107311] [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: 01/10/2024] [Revised: 04/12/2024] [Accepted: 04/16/2024] [Indexed: 04/26/2024] Open
Abstract
The Hippo signaling pathway plays an essential role in organ size control and tumorigenesis. Loss of Hippo signal and hyper-activation of the downstream oncogenic YAP signaling are commonly observed in various types of cancers. We previously identified STRN3-containing PP2A phosphatase as a negative regulator of MST1/2 kinases (i.e., Hippo) in gastric cancer (GC), opening the possibility of selectively targeting the PP2Aa-STRN3-MST1/2 axis to recover Hippo signaling against cancer. Here, we further discovered 1) disulfiram (DSF), an FDA-approved drug, which can similarly block the binding of STRN3 to PP2A core enzyme and 2) CX-6258 (CX), a chemical inhibitor, that can disrupt the interaction between STRN3 and MST1/2, both allowing reactivation of Hippo activity to inhibit GC. More importantly, we found these two compounds, via an MST1/2 kinase-dependent manner, inhibit DNA repair to sensitize GC towards chemotherapy. In addition, we identified thiram, a structural analog of DSF, can function similarly to inhibit cancer cell proliferation or enhance chemotherapy sensitivity. Interestingly, inclusion of copper ion enhanced such effects of DSF and thiram on GC treatment. Overall, this work demonstrated that pharmacological targeting of the PP2Aa-STRN3-MST1/2 axis by drug compounds can potently recover Hippo signal for tumor treatment.
Collapse
Affiliation(s)
- Zhifa Cao
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, China; Department of Stomatology, Department of Medical Ultrasound, Shanghai Tenth People's Hospital, Tongji University Cancer Center, School of Medicine, Tongji University, Shanghai, China
| | - Yu Hou
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhangting Zhao
- Department of Stomatology, Department of Medical Ultrasound, Shanghai Tenth People's Hospital, Tongji University Cancer Center, School of Medicine, Tongji University, Shanghai, China
| | - Hui Zhang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Luyang Tian
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yiming Zhang
- Department of Stomatology, Department of Medical Ultrasound, Shanghai Tenth People's Hospital, Tongji University Cancer Center, School of Medicine, Tongji University, Shanghai, China
| | - Chao Dong
- Department of Medical Oncology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Fenghua Guo
- Department of General Surgery, Hua'shan Hospital, Fudan University Shanghai Medical College, Shanghai, China
| | - Lijie Tan
- Department of Thoracic Surgery, Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yi Han
- Department of Stomatology, Department of Medical Ultrasound, Shanghai Tenth People's Hospital, Tongji University Cancer Center, School of Medicine, Tongji University, Shanghai, China
| | - Wenjia Wang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shi Jiao
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yang Tang
- Department of Stomatology, Department of Medical Ultrasound, Shanghai Tenth People's Hospital, Tongji University Cancer Center, School of Medicine, Tongji University, Shanghai, China.
| | - Liwei An
- Department of Stomatology, Department of Medical Ultrasound, Shanghai Tenth People's Hospital, Tongji University Cancer Center, School of Medicine, Tongji University, Shanghai, China.
| | - Zhaocai Zhou
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, China; Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
8
|
Lin G, Xia A, Qiao J, Zhang H, Chen P, Zhou P, Hu Q, Xiang Z, Zhang S, Li L, Yang S. Identification of a new class of activators of the Hippo pathway with antitumor activity in vitro and in vivo. Biochem Pharmacol 2024; 224:116217. [PMID: 38641306 DOI: 10.1016/j.bcp.2024.116217] [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: 11/24/2023] [Revised: 03/05/2024] [Accepted: 04/15/2024] [Indexed: 04/21/2024]
Abstract
The Hippo pathway is a key regulator of tissue growth, organ size, and tumorigenesis. Activating the Hippo pathway by gene editing or pharmaceutical intervention has been proven to be a new therapeutic strategy for treatment of the Hippo pathway-dependent cancers. To now, a number of compounds that directly target the downstream effector proteins of Hippo pathway, including YAP and TEADs, have been disclosed, but very few Hippo pathway activators are reported. Here, we discovered a new class of Hippo pathway activator, YL-602, which inhibited CTGF expression in cells irrespective of cell density and the presence of serum. Mechanistically, YL-602 activates the Hippo pathway via MST1/2, which is different from known activators of Hippo pathway. In vitro, YL-602 significantly induced tumor cell apoptosis and inhibited colony formation of tumor cells. In vivo, oral administration of YL-602 substantially suppressed the growth of cancer cells by activation of Hippo pathway. Overall, YL-602 could be a promising lead compound, and deserves further investigation for its mechanism of action and therapeutic applications.
Collapse
Affiliation(s)
- Guifeng Lin
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China; Fujian Key Laboratory of Natural Medicine Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Anjie Xia
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China; Department of Ophthalmology and Research Laboratory of Macular Disease, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jingxin Qiao
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hailin Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Pei Chen
- Key Laboratory of Drug Targeting and Drug Delivery System of Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan 610041, China
| | - Pei Zhou
- Key Laboratory of Drug Targeting and Drug Delivery System of Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan 610041, China
| | - Qian Hu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhiyu Xiang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Shiyu Zhang
- Key Laboratory of Drug Targeting and Drug Delivery System of Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan 610041, China
| | - Linli Li
- Key Laboratory of Drug Targeting and Drug Delivery System of Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan 610041, China.
| | - Shengyong Yang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
9
|
Xue Q, Yan Y, Zhang K, Zhang H, Zhao Y. Exposure to microcystin-LR promotes astrocyte proliferation both in vitro and in vivo via Hippo signaling pathway. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 279:116480. [PMID: 38772146 DOI: 10.1016/j.ecoenv.2024.116480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 05/14/2024] [Accepted: 05/16/2024] [Indexed: 05/23/2024]
Abstract
Microcystins (MCs) are toxic to the central nervous system of mammals. However, the direct toxicity of MCs on mammalian brain cells and the involved molecular mechanisms are not fully elucidated. Here, we incubated primary astrocytes, the major glial cell-type in the brain, with 0-12.5 μM concentrations of MC-LR for 48 h, and the impairment was evaluated. We found that MC-LR caused significant increases in the cell viability at the range of 0.05-1 μM concentrations with the highest density at 0.1 μM concentration. Treatment with 0.1 μM MC-LR induced YAP nuclear translocation and decreased the ratio of p-YAP to YAP. It also decreased mRNA levels of the upstream regulator (AMOT), and enhanced expressions of YAP interacted genes (Egfr, Tead1, and Ctgf) in primary astrocytes. Overexpression of AMOT significantly attenuated the increase of MC-LR-induced astrocyte proliferation and the expression of YAP downstream genes. These results indicate that Hippo signaling contributed to MC-LR-caused astrocyte proliferation. Further, reactive astrogliosis was observed in the mice brain after MC-LR exposure to environmentally relevant concentrations (20 or 100 μg/L) through drinking water for 16 weeks. Pathological observations revealed that 100 μg/L MC-LR exposure caused neuronal damages with characteristics of shrunken or vacuolation in the region of the cerebral cortex, striatum and cerebellum. These results were accompanied with increased oxidative stress and inflammatory response. Our data reveal the potential astrocytic mechanisms in MC-induced neurotoxicity and raise an alarm for neurodegenerative disease risk following daily exposure to MC-LR.
Collapse
Affiliation(s)
- Qingju Xue
- State Key Laboratory of Lake Science and Environment, Nanjing Institute of Geography and Limnology, Chinese Academy of Sciences, 73 East Beijing Road, Nanjing 210008, PR China
| | - Yunjun Yan
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, PR China
| | - Kaiye Zhang
- State Key Laboratory of Lake Science and Environment, Nanjing Institute of Geography and Limnology, Chinese Academy of Sciences, 73 East Beijing Road, Nanjing 210008, PR China
| | - Hui Zhang
- State Key Laboratory of Lake Science and Environment, Nanjing Institute of Geography and Limnology, Chinese Academy of Sciences, 73 East Beijing Road, Nanjing 210008, PR China
| | - Yanyan Zhao
- State Key Laboratory of Lake Science and Environment, Nanjing Institute of Geography and Limnology, Chinese Academy of Sciences, 73 East Beijing Road, Nanjing 210008, PR China.
| |
Collapse
|
10
|
Song Y, Na H, Lee SE, Kim YM, Moon J, Nam TW, Ji Y, Jin Y, Park JH, Cho SC, Lee J, Hwang D, Ha SJ, Park HW, Kim JB, Lee HW. Dysfunctional adipocytes promote tumor progression through YAP/TAZ-dependent cancer-associated adipocyte transformation. Nat Commun 2024; 15:4052. [PMID: 38744820 PMCID: PMC11094189 DOI: 10.1038/s41467-024-48179-3] [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: 06/19/2023] [Accepted: 04/23/2024] [Indexed: 05/16/2024] Open
Abstract
Obesity has emerged as a prominent risk factor for the development of malignant tumors. However, the existing literature on the role of adipocytes in the tumor microenvironment (TME) to elucidate the correlation between obesity and cancer remains insufficient. Here, we aim to investigate the formation of cancer-associated adipocytes (CAAs) and their contribution to tumor growth using mouse models harboring dysfunctional adipocytes. Specifically, we employ adipocyte-specific BECN1 KO (BaKO) mice, which exhibit lipodystrophy due to dysfunctional adipocytes. Our results reveal the activation of YAP/TAZ signaling in both CAAs and BECN1-deficient adipocytes, inducing adipocyte dedifferentiation and formation of a malignant TME. The additional deletion of YAP/TAZ from BaKO mice significantly restores the lipodystrophy and inflammatory phenotypes, leading to tumor regression. Furthermore, mice fed a high-fat diet (HFD) exhibit decreased BECN1 and increased YAP/TAZ expression in their adipose tissues. Treatment with the YAP/TAZ inhibitor, verteporfin, suppresses tumor progression in BaKO and HFD-fed mice, highlighting its efficacy against mice with metabolic dysregulation. Overall, our findings provide insights into the key mediators of CAA and their significance in developing a TME, thereby suggesting a viable approach targeting adipocyte homeostasis to suppress cancer growth.
Collapse
Affiliation(s)
- Yaechan Song
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Heeju Na
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Seung Eon Lee
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - You Min Kim
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Jihyun Moon
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Tae Wook Nam
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Yul Ji
- Department of Biological Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Young Jin
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Jae Hyung Park
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Seok Chan Cho
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Jaehoon Lee
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
- Gemcro, Inc, Seoul, 03722, Republic of Korea
| | - Daehee Hwang
- Department of Biological Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Sang-Jun Ha
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Hyun Woo Park
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Jae Bum Kim
- Department of Biological Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Han-Woong Lee
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea.
- Gemcro, Inc, Seoul, 03722, Republic of Korea.
| |
Collapse
|
11
|
Ryu HJ, Kim C, Jang H, Kim SI, Shin SJ, Chung KY, Torres-Cabala C, Kim SK. Nuclear Localization of Yes-Associated Protein Is Associated With Tumor Progression in Cutaneous Melanoma. J Transl Med 2024; 104:102048. [PMID: 38490470 DOI: 10.1016/j.labinv.2024.102048] [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: 10/17/2023] [Revised: 02/26/2024] [Accepted: 03/07/2024] [Indexed: 03/17/2024] Open
Abstract
Yes-associated protein (YAP), an effector molecule of the Hippo signaling pathway, is expressed at high levels in cutaneous melanoma. However, the role of YAP in melanoma progression according to cellular localization is poorly understood. Tissues from 140 patients with invasive melanoma were evaluated by immunohistochemistry. Flow cytometry, western blotting, viability assays, wound healing assays, verteporfin treatment, and xenograft assays were conducted using melanoma cell lines B16F1 and B16F10 subjected to YapS127A transfection and siYap knockdown. Nuclear YAP localization was identified in 63 tumors (45.0%) and was more frequent than cytoplasmic YAP in acral lentiginous and nodular subtypes (P = .007). Compared with cytoplasmic YAP melanomas, melanomas with nuclear YAP had higher mitotic activity (P = .016), deeper invasion (P < .001), and more frequently metastasized to lymph nodes (P < .001) and distant organs (P < .001). Patients with nuclear YAP melanomas had poorer disease-free survival (P < .001) and overall survival (P < .001). Nuclear YAP was an independent risk factor for distant metastasis (hazard ratio: 3.206; 95% CI, 1.032-9.961; P = .044). Proliferative ability was decreased in siYapB16F1 (P < .001) and siYapB16F10 (P = .001) cells and increased in YapS127AB16F1 (P = .003) and YapS127AB16F10 (P = .002) cells. Cell cycle analysis demonstrated relative G1 retention in siYapB16F1 (P < .001) and siYapB16F10 (P < .001) cells and S retention in YapS127AB16F1 cells (P = .008). Wound healing assays showed that Yap knockdown inhibited cell invasion (siYapB16F1, P = .001; siYapB16F10, P < .001), whereas nuclear YAP promoted it (YapS127AB16F, P < .001; YapS127AB16F1, P = .017). Verteporfin, a direct YAP inhibitor, reduced cellular proliferation in B16F1 (P = .003) and B16F10 (P < .001) cells. Proliferative effects of nuclear YAP were confirmed in xenograft mice (P < .001). In conclusion, nuclear YAP in human melanomas showed subtype specificity and correlated with proliferative activity and proinvasiveness. It is expected that YAP becomes a useful prognostic marker, and its inhibition may be a potential therapy for melanoma patients.
Collapse
Affiliation(s)
- Hyang Joo Ryu
- Department of Pathology, Yonsei University College of Medicine, Seoul, South Korea
| | - Chayeon Kim
- Department of Pathology, Yonsei University College of Medicine, Seoul, South Korea
| | - Hyenguk Jang
- Department of Pathology, Yonsei University College of Medicine, Seoul, South Korea
| | - Sun Il Kim
- Department of Pathology, Yonsei University College of Medicine, Seoul, South Korea
| | - Sang Joon Shin
- Department of Oncology, Yonsei University College of Medicine, Seoul, South Korea
| | - Kee Yang Chung
- Department of Dermatology, Yonsei University College of Medicine, Seoul, South Korea
| | - Carlos Torres-Cabala
- Department of Pathology, The University of Texas, MD Anderson Cancer Center, Houston, Texas.
| | - Sang Kyum Kim
- Department of Pathology, Yonsei University College of Medicine, Seoul, South Korea.
| |
Collapse
|
12
|
Qian H, Ding CH, Liu F, Chen SJ, Huang CK, Xiao MC, Hong XL, Wang MC, Yan FZ, Ding K, Cui YL, Zheng BN, Ding J, Luo C, Zhang X, Xie WF. SRY-Box transcription factor 9 triggers YAP nuclear entry via direct interaction in tumors. Signal Transduct Target Ther 2024; 9:96. [PMID: 38653754 PMCID: PMC11039692 DOI: 10.1038/s41392-024-01805-4] [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: 08/21/2023] [Revised: 01/27/2024] [Accepted: 03/13/2024] [Indexed: 04/25/2024] Open
Abstract
The translocation of YAP from the cytoplasm to the nucleus is critical for its activation and plays a key role in tumor progression. However, the precise molecular mechanisms governing the nuclear import of YAP are not fully understood. In this study, we have uncovered a crucial role of SOX9 in the activation of YAP. SOX9 promotes the nuclear translocation of YAP by direct interaction. Importantly, we have identified that the binding between Asp-125 of SOX9 and Arg-124 of YAP is essential for SOX9-YAP interaction and subsequent nuclear entry of YAP. Additionally, we have discovered a novel asymmetrical dimethylation of YAP at Arg-124 (YAP-R124me2a) catalyzed by PRMT1. YAP-R124me2a enhances the interaction between YAP and SOX9 and is associated with poor prognosis in multiple cancers. Furthermore, we disrupted the interaction between SOX9 and YAP using a competitive peptide, S-A1, which mimics an α-helix of SOX9 containing Asp-125. S-A1 significantly inhibits YAP nuclear translocation and effectively suppresses tumor growth. This study provides the first evidence of SOX9 as a pivotal regulator driving YAP nuclear translocation and presents a potential therapeutic strategy for YAP-driven human cancers by targeting SOX9-YAP interaction.
Collapse
Affiliation(s)
- Hui Qian
- Department of Gastroenterology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Chen-Hong Ding
- Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Fang Liu
- Department of Gastroenterology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Shi-Jie Chen
- Drug Discovery and Design Center, CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Chen-Kai Huang
- Department of Gastroenterology, First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Meng-Chao Xiao
- Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xia-Lu Hong
- Department of Gastroenterology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Ming-Chen Wang
- Drug Discovery and Design Center, CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Fang-Zhi Yan
- Department of Gastroenterology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Kai Ding
- Department of Gastroenterology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Ya-Lu Cui
- Department of Gastroenterology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Bai-Nan Zheng
- Department of Gastroenterology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Jin Ding
- Clinical Cancer Institute, Center for Translational Medicine, Naval Medical University, Shanghai, China
| | - Cheng Luo
- Drug Discovery and Design Center, CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
| | - Xin Zhang
- Department of Gastroenterology, Changzheng Hospital, Naval Medical University, Shanghai, China.
| | - Wei-Fen Xie
- Department of Gastroenterology, Changzheng Hospital, Naval Medical University, Shanghai, China.
| |
Collapse
|
13
|
Huang HC, Wang TY, Rousseau J, Orlando M, Mungaray M, Michaud C, Plaisier C, Chen ZB, Wang KC. Biomimetic nanodrug targets inflammation and suppresses YAP/TAZ to ameliorate atherosclerosis. Biomaterials 2024; 306:122505. [PMID: 38359507 PMCID: PMC11479593 DOI: 10.1016/j.biomaterials.2024.122505] [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: 05/14/2023] [Revised: 01/21/2024] [Accepted: 02/07/2024] [Indexed: 02/17/2024]
Abstract
Atherosclerosis, a chronic inflammatory disease, is the primary cause of myocardial infarction and ischemic stroke. Recent studies have demonstrated that dysregulation of yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding domain (TAZ) contributes to plaque development, making YAP/TAZ potential therapeutic targets. However, systemic modulation of YAP/TAZ expression or activities risks serious off-target effects, limiting clinical applicability. To address the challenge, this study develops monocyte membrane-coated nanoparticles (MoNP) as a targeted delivery system for activated and inflamed endothelium lining the plaque surface. The MoNP system is used to deliver verteporfin (VP), aimed at inhibiting YAP/TAZ specifically within arterial regions prone to atherosclerosis. The results reveal that MoNP significantly enhance payload delivery to inflamed endothelial cells (EC) while avoiding phagocytic cells. When administered in mice, MoNP predominantly accumulate in intima of the atheroprone artery. MoNP-mediated delivery of VP substantially reduces YAP/TAZ expression, thereby suppressing inflammatory gene expression and macrophage infiltration in cultured EC and mouse arteries exposed to atherogenic stimuli. Importantly, this targeted VP nanodrug effectively decreases plaque development in mice without causing noticeable histopathological changes in major organs. Collectively, these findings demonstrate a lesion-targeted and pathway-specific biomimetic nanodrug, potentially leading to safer and more effective treatments for atherosclerosis.
Collapse
Affiliation(s)
- Hui-Chun Huang
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, 85287, USA
| | - Ting-Yun Wang
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, 85287, USA; School for Engineering of Matter, Transport and Energy, Arizona State University, Tempe, AZ, 85287, USA
| | - Joshua Rousseau
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, 85287, USA
| | - Mark Orlando
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, 85287, USA
| | - Michelle Mungaray
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, 85287, USA
| | - Chamonix Michaud
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, 85287, USA
| | - Christopher Plaisier
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, 85287, USA
| | - Zhen Bouman Chen
- Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope, Duarte, CA, 91010, USA
| | - Kuei-Chun Wang
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, 85287, USA.
| |
Collapse
|
14
|
Farris F, Elhagh A, Vigorito I, Alongi N, Pisati F, Giannattasio M, Casagrande F, Veghini L, Corbo V, Tripodo C, Di Napoli A, Matafora V, Bachi A. Unveiling the mechanistic link between extracellular amyloid fibrils, mechano-signaling and YAP activation in cancer. Cell Death Dis 2024; 15:28. [PMID: 38199984 PMCID: PMC10781709 DOI: 10.1038/s41419-024-06424-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: 07/25/2023] [Revised: 12/20/2023] [Accepted: 01/02/2024] [Indexed: 01/12/2024]
Abstract
The tumor microenvironment is a complex ecosystem that plays a critical role in cancer progression and treatment response. Recently, extracellular amyloid fibrils have emerged as novel components of the tumor microenvironment; however, their function remains elusive. In this study, we establish a direct connection between the presence of amyloid fibrils in the secretome and the activation of YAP, a transcriptional co-activator involved in cancer proliferation and drug resistance. Furthermore, we uncover a shared mechano-signaling mechanism triggered by amyloid fibrils in both melanoma and pancreatic ductal adenocarcinoma cells. Our findings highlight the crucial role of the glycocalyx protein Agrin which binds to extracellular amyloid fibrils and acts as a necessary factor in driving amyloid-dependent YAP activation. Additionally, we reveal the involvement of the HIPPO pathway core kinase LATS1 in this signaling cascade. Finally, we demonstrate that extracellular amyloid fibrils enhance cancer cell migration and invasion. In conclusion, our research expands our knowledge of the tumor microenvironment by uncovering the role of extracellular amyloid fibrils in driving mechano-signaling and YAP activation. This knowledge opens up new avenues for developing innovative strategies to modulate YAP activation and mitigate its detrimental effects during cancer progression.
Collapse
Affiliation(s)
- Francesco Farris
- IFOM ETS - The AIRC Institute of Molecular Oncology, 20139, Milan, Italy
| | - Alice Elhagh
- IFOM ETS - The AIRC Institute of Molecular Oncology, 20139, Milan, Italy
| | - Ilaria Vigorito
- IFOM ETS - The AIRC Institute of Molecular Oncology, 20139, Milan, Italy
| | - Nicoletta Alongi
- IFOM ETS - The AIRC Institute of Molecular Oncology, 20139, Milan, Italy
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Federica Pisati
- Histopathology Unit, Cogentech S.C.a.R.L, 20139, Milan, Italy
| | - Michele Giannattasio
- IFOM ETS - The AIRC Institute of Molecular Oncology, 20139, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, 20122, Milan, Italy
| | - Francesca Casagrande
- IFOM ETS - The AIRC Institute of Molecular Oncology, 20139, Milan, Italy
- Human Technopole, Milan, Italy
| | - Lisa Veghini
- Department of Engineering for Innovation Medicine (DIMI), University of Verona, 37134, Verona, Italy
| | - Vincenzo Corbo
- Department of Engineering for Innovation Medicine (DIMI), University of Verona, 37134, Verona, Italy
- ARC-Net Centre for Applied Research on Cancer, University of Verona, 37134, Verona, Italy
| | - Claudio Tripodo
- IFOM ETS - The AIRC Institute of Molecular Oncology, 20139, Milan, Italy
- Tumor Immunology Unit, Department of Health Sciences, University of Palermo, 90133, Palermo, Italy
| | - Arianna Di Napoli
- Pathology Unit, Department of Clinical and Molecular Medicine, Sant'Andrea University Hospital, Sapienza University of Rome, 00189, Rome, Italy
| | - Vittoria Matafora
- IFOM ETS - The AIRC Institute of Molecular Oncology, 20139, Milan, Italy.
| | - Angela Bachi
- IFOM ETS - The AIRC Institute of Molecular Oncology, 20139, Milan, Italy.
| |
Collapse
|
15
|
Ma F, Tsou PS, Gharaee-Kermani M, Plazyo O, Xing X, Kirma J, Wasikowski R, Hile GA, Harms PW, Jiang Y, Xing E, Nakamura M, Ochocki D, Brodie WD, Pillai S, Maverakis E, Pellegrini M, Modlin RL, Varga J, Tsoi LC, Lafyatis R, Kahlenberg JM, Billi AC, Khanna D, Gudjonsson JE. Systems-based identification of the Hippo pathway for promoting fibrotic mesenchymal differentiation in systemic sclerosis. Nat Commun 2024; 15:210. [PMID: 38172207 PMCID: PMC10764940 DOI: 10.1038/s41467-023-44645-6] [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: 02/27/2023] [Accepted: 12/23/2023] [Indexed: 01/05/2024] Open
Abstract
Systemic sclerosis (SSc) is a devastating autoimmune disease characterized by excessive production and accumulation of extracellular matrix, leading to fibrosis of skin and other internal organs. However, the main cellular participants in SSc skin fibrosis remain incompletely understood. Here using differentiation trajectories at a single cell level, we demonstrate a dual source of extracellular matrix deposition in SSc skin from both myofibroblasts and endothelial-to-mesenchymal-transitioning cells (EndoMT). We further define a central role of Hippo pathway effectors in differentiation and homeostasis of myofibroblast and EndoMT, respectively, and show that myofibroblasts and EndoMTs function as central communication hubs that drive key pro-fibrotic signaling pathways in SSc. Together, our data help characterize myofibroblast differentiation and EndoMT phenotypes in SSc skin, and hint that modulation of the Hippo pathway may contribute in reversing the pro-fibrotic phenotypes in myofibroblasts and EndoMTs.
Collapse
Affiliation(s)
- Feiyang Ma
- Department of Dermatology, University of Michigan, Ann Arbor, MI, USA
- Division of Rheumatology, Dept of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Pei-Suen Tsou
- Division of Rheumatology, Dept of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- University of Michigan Scleroderma Program, Ann Arbor, MI, USA
| | - Mehrnaz Gharaee-Kermani
- Division of Rheumatology, Dept of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Olesya Plazyo
- Division of Rheumatology, Dept of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Xianying Xing
- Division of Rheumatology, Dept of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Joseph Kirma
- Division of Rheumatology, Dept of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Rachael Wasikowski
- Division of Rheumatology, Dept of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Grace A Hile
- Division of Rheumatology, Dept of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Paul W Harms
- Department of Dermatology, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Yanyun Jiang
- Department of Dermatology, University of Michigan, Ann Arbor, MI, USA
| | - Enze Xing
- Division of Rheumatology, Dept of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Mio Nakamura
- Department of Dermatology, University of Michigan, Ann Arbor, MI, USA
| | - Danielle Ochocki
- Division of Rheumatology, Dept of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- University of Michigan Scleroderma Program, Ann Arbor, MI, USA
| | - William D Brodie
- Division of Rheumatology, Dept of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Shiv Pillai
- Ragon Institute, Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Boston, MA, USA
| | - Emanual Maverakis
- Department of Dermatology, University of California, Davis, Sacramento, CA, USA
| | - Matteo Pellegrini
- Dept of Molecular, Cell, and Developmental Biology, University of California Los Angeles, Los Angeles, CA, USA
| | - Robert L Modlin
- Dept of Molecular, Cell, and Developmental Biology, University of California Los Angeles, Los Angeles, CA, USA
- Division of Dermatology, Department of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - John Varga
- Division of Rheumatology, Dept of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- University of Michigan Scleroderma Program, Ann Arbor, MI, USA
| | - Lam C Tsoi
- Department of Dermatology, University of Michigan, Ann Arbor, MI, USA
| | - Robert Lafyatis
- Division of Rheumatology, University of Pittsburgh, Pittsburgh, PA, USA
| | - J Michelle Kahlenberg
- Division of Rheumatology, Dept of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Allison C Billi
- Department of Dermatology, University of Michigan, Ann Arbor, MI, USA
| | - Dinesh Khanna
- Division of Rheumatology, Dept of Internal Medicine, University of Michigan, Ann Arbor, MI, USA.
- University of Michigan Scleroderma Program, Ann Arbor, MI, USA.
| | | |
Collapse
|
16
|
Li H, Zou F, Zhang J, Zhu S, Chu K, Zhang X, Zhao T. YAP suppresses human T-cell leukemia virus type 1 transcription. J Med Virol 2023; 95:e29065. [PMID: 37661566 DOI: 10.1002/jmv.29065] [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: 07/17/2023] [Revised: 08/11/2023] [Accepted: 08/17/2023] [Indexed: 09/05/2023]
Abstract
Human T-cell leukemia virus type 1 (HTLV-1) is an oncogenic retrovirus that causes adult T-cell leukemia/lymphoma (ATL). HTLV-1 encodes Tax protein that activates transcription from viral long terminal repeats (LTR). Multiple cofactors are involved in the regulation of HTLV-1 transcription via association with Tax. Yes-associated protein (YAP), which is the key effector of Hippo pathway, is elevated and activated in ATL cells. In this study, we reported that YAP protein suppressed Tax activation of HTLV-1 5' LTR but not 3' LTR. The activation of the 5' LTR by Tax was potentiated when YAP was depleted. Moreover, overexpression of YAP repressed HTLV-1 plus-strand viral gene expression and virion production, whereas compromising YAP by RNA inference augmented the expression of HTLV-1 protein. As mechanisms of YAP-mediated viral transcription inhibition, we found that YAP interacted with Tax, and prevented the association between Tax and p300. It finally led to the inhibition of recruitment of Tax to the Tax-responsive element in the 5' LTR of HTLV-1. Taken together, our results demonstrate the negative regulatory function of YAP in Tax activation of HTLV-1 transcription. It may achieve sufficient transcriptional repression to maintain persistent infection and long-term latency of HTLV-1 in the host cells.
Collapse
Affiliation(s)
- Hengbo Li
- School of Medicine, Hangzhou City University, Hangzhou, China
- College of Life Sciences, Zhejiang Normal University, Jinhua, China
| | - Feng Zou
- College of Life Sciences, Zhejiang Normal University, Jinhua, China
| | - Jie Zhang
- School of Medicine, Hangzhou City University, Hangzhou, China
| | - Shengyu Zhu
- School of Medicine, Hangzhou City University, Hangzhou, China
| | - Kaifei Chu
- School of Medicine, Hangzhou City University, Hangzhou, China
- College of Life Sciences, Zhejiang Normal University, Jinhua, China
| | - Xu Zhang
- School of Medicine, Hangzhou City University, Hangzhou, China
- College of Life Sciences, Zhejiang Normal University, Jinhua, China
| | - Tiejun Zhao
- School of Medicine, Hangzhou City University, Hangzhou, China
- College of Life Sciences, Zhejiang Normal University, Jinhua, China
| |
Collapse
|
17
|
Bertagnin C, Messa L, Pavan M, Celegato M, Sturlese M, Mercorelli B, Moro S, Loregian A. A small molecule targeting the interaction between human papillomavirus E7 oncoprotein and cellular phosphatase PTPN14 exerts antitumoral activity in cervical cancer cells. Cancer Lett 2023; 571:216331. [PMID: 37532093 DOI: 10.1016/j.canlet.2023.216331] [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: 04/27/2023] [Revised: 07/21/2023] [Accepted: 07/30/2023] [Indexed: 08/04/2023]
Abstract
Human papillomavirus (HPV)-induced cancers still represent a major health issue for worldwide population and lack specific therapeutic regimens. Despite substantial advancements in anti-HPV vaccination, the incidence of HPV-related cancers remains high, thus there is an urgent need for specific anti-HPV drugs. The HPV E7 oncoprotein is a major driver of carcinogenesis that acts by inducing the degradation of several host factors. A target is represented by the cellular phosphatase PTPN14 and its E7-mediated degradation was shown to be crucial in HPV oncogenesis. Here, by exploiting the crystal structure of E7 bound to PTPN14, we performed an in silico screening of small-molecule compounds targeting the C-terminal CR3 domain of E7 involved in the interaction with PTPN14. We discovered a compound able to inhibit the E7/PTPN14 interaction in vitro and to rescue PTPN14 levels in cells, leading to a reduction in viability, proliferation, migration, and cancer-stem cell potential of HPV-positive cervical cancer cells. Mechanistically, as a consequence of PTPN14 rescue, treatment of cancer cells with this compound altered the Yes-associated protein (YAP) nuclear-cytoplasmic shuttling and downstream signaling. Notably, this compound was active against cervical cancer cells transformed by different high-risk (HR)-HPV genotypes indicating a potential broad-spectrum activity. Overall, our study reports the first-in-class inhibitor of E7/PTPN14 interaction and provides the proof-of-principle that pharmacological inhibition of this interaction by small-molecule compounds could be a feasible therapeutic strategy for the development of novel antitumoral drugs specific for HPV-associated cancers.
Collapse
Affiliation(s)
- Chiara Bertagnin
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | - Lorenzo Messa
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | - Matteo Pavan
- Molecular Modeling Section (MMS), Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy
| | - Marta Celegato
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | - Mattia Sturlese
- Molecular Modeling Section (MMS), Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy
| | | | - Stefano Moro
- Molecular Modeling Section (MMS), Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy
| | - Arianna Loregian
- Department of Molecular Medicine, University of Padua, Padua, Italy.
| |
Collapse
|
18
|
Osama M, Essibayi MA, Osama M, Ibrahim IA, Nasr Mostafa M, Şakir Ekşi M. The impact of interaction between verteporfin and yes-associated protein 1/transcriptional coactivator with PDZ-binding motif-TEA domain pathway on the progression of isocitrate dehydrogenase wild-type glioblastoma. J Cent Nerv Syst Dis 2023; 15:11795735231195760. [PMID: 37600236 PMCID: PMC10439684 DOI: 10.1177/11795735231195760] [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: 01/12/2023] [Accepted: 08/02/2023] [Indexed: 08/22/2023] Open
Abstract
Verteporfin and 5-ALA are used for visualizing malignant tissue components in different body tumors and as photodynamic therapy in treating isocitrate dehydrogenase (IDH) wild-type glioblastoma (GBM). Additionally, verteporfin interferes with Yes-associated protein 1 (YAP)/Transcriptional coactivator with PDZ-binding motif - TEA domain (TAZ-TEAD) pathway, thus inhibiting the downstream effect of these oncogenes and reducing the malignant properties of GBM. Animal studies have shown verteporfin to be successful in increasing survival rates, which have led to the conduction of phase 1 and 2 clinical trials to further investigate its efficacy in treating GBM. In this article, we aimed to review the novel mechanism of verteporfin's action, the impact of its interaction with YAP/TAZ-TEAD, its effect on glioblastoma stem cells, and its role in inducing ferroptosis.
Collapse
Affiliation(s)
- Mahmoud Osama
- Department of Neurosurgery, Nasser Institute for Research and Treatment, Cairo, Egypt
- Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Muhammed Amir Essibayi
- Department of Neurosurgery, Albert Einstein College of Medicine, New York City, NY, USA
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - Mona Osama
- Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Ismail A. Ibrahim
- Department of Physical Therapy and Rehabilitation, Fenerbahce University, Istanbul, Turkey
| | | | - Murat Şakir Ekşi
- Neurosurgery Clinic, FSM Training and Research Hospital, Istanbul, Turkey
| |
Collapse
|
19
|
Wang XW, Zhao R, Yang ZY, Li T, Yang JC, Wang XL, Li XT, Zhao XR, Li XZ, Wang XX. YAP inhibitor verteporfin suppresses tumor angiogenesis and overcomes chemoresistance in esophageal squamous cell carcinoma. J Cancer Res Clin Oncol 2023; 149:7703-7716. [PMID: 37000262 DOI: 10.1007/s00432-023-04722-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 03/22/2023] [Indexed: 04/01/2023]
Abstract
PURPOSE Targeting angiogenesis is an attractive strategy for the effective treatment of cancer. This study aimed to investigate the anti-cancer activities of YAP inhibitor verteporfin (VP) in esophageal squamous cell carcinoma (ESCC) cells through its inhibitory effect on tumor angiogenesis. METHODS Cell proliferation, apoptosis, migration and invasion abilities were estimated by MTT, colony formation, DAPI staining, wound healing and transwell assays, respectively. Human umbilical vein endothelial cell (HUVEC) tube formation assay and chick embryo chorioallantoic membrane (CAM) model were used to observe angiogenesis in vitro and in vivo. The interactions between ESCC cells and HUVECs were assessed by cell chemotactic migration and adhesion assays. The expression levels of angiogenesis-related molecules were detected by Western blot. RESULTS We found that VP was potential to inhibit ESCC cell proliferation, migration, invasion and induce apoptosis in the dose-dependent fashion. VP also significantly suppressed proliferation, migration, and tube formation of HUVECs and promoted apoptosis of HUVECs, and reduced angiogenesis in CAM. Moreover, VP inhibited ESCC cell-induced angiogenesis in vitro by decreasing HUVEC chemotactic migration, adhesion and tube formation, and also reduced ESCC cell-induced neovascularization of the CAM in vivo. In addition, VP suppressed the expression of pro-angiogenic molecules such as VEGFA, MMP-2 and β-catenin in ESCC cells. Furtherly, VP increased the chemosensitivity of ESCC-resistant cells to paclitaxel (PTX). The combination of VP and PTX attenuated the resistant cell-mediated angiogenesis in vitro and in vivo. CONCLUSION These results reveal for the first time that VP potently inhibits malignant progression and overcomes chemoresistance of ESCC cells via inhibition of tumor angiogenesis. It provides insight into a new strategy for the treatment of ESCC that VP could be a potential drug candidate for targeting tumor angiogenesis.
Collapse
Affiliation(s)
- Xue-Wei Wang
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, China
| | - Rong Zhao
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, China
| | - Zi-Yi Yang
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, China
| | - Ting Li
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, China
| | - Jia-Cheng Yang
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, China
| | - Xiu-Li Wang
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, China
| | - Xin-Ting Li
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, China
| | - Xin-Ran Zhao
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, China
| | - Xiao-Zhong Li
- Department of Infectious Diseases, Shanxi Provincial People's Hospital, Taiyuan, China
| | - Xiao-Xia Wang
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, China.
| |
Collapse
|
20
|
Clevenger AJ, McFarlin MK, Collier CA, Sheshadri VS, Madyastha AK, Gorley JPM, Solberg SC, Stratman AN, Raghavan SA. Peristalsis-Associated Mechanotransduction Drives Malignant Progression of Colorectal Cancer. Cell Mol Bioeng 2023; 16:261-281. [PMID: 37811008 PMCID: PMC10550901 DOI: 10.1007/s12195-023-00776-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 07/21/2023] [Indexed: 10/10/2023] Open
Abstract
Introduction In the colorectal cancer (CRC) tumor microenvironment, cancerous and precancerous cells continuously experience mechanical forces associated with peristalsis. Given that mechanical forces like shear stress and strain can positively impact cancer progression, we explored the hypothesis that peristalsis may also contribute to malignant progression in CRC. We defined malignant progression as enrichment of cancer stem cells and the acquisition of invasive behaviors, both vital to CRC progression. Methods We leveraged our peristalsis bioreactor to expose CRC cell lines (HCT116), patient-derived xenograft (PDX1,2) lines, or non-cancerous intestinal cells (HIEC-6) to forces associated with peristalsis in vitro. Cells were maintained in static control conditions or exposed to peristalsis for 24 h prior to assessment of cancer stem cell (CSC) emergence or the acquisition of invasive phenotypes. Results Exposure of HCT116 cells to peristalsis significantly increased the emergence of LGR5+ CSCs by 1.8-fold compared to static controls. Peristalsis enriched LGR5 positivity in several CRC cell lines, notably significant in KRAS mutant lines. In contrast, peristalsis failed to increase LGR5+ in non-cancerous intestinal cells, HIEC-6. LGR5+ emergence downstream of peristalsis was dependent on ROCK and Wnt activity, and not YAP1 activation. Additionally, HCT116 cells adopted invasive morphologies when exposed to peristalsis, with increased filopodia density and epithelial to mesenchymal gene expression, in a Wnt dependent manner. Conclusions Peristalsis associated forces drive malignant progression of CRC via ROCK, YAP1, and Wnt-related mechanotransduction. Supplementary Information The online version contains supplementary material available at 10.1007/s12195-023-00776-w.
Collapse
Affiliation(s)
- Abigail J. Clevenger
- Department of Biomedical Engineering, Texas A&M University, 5016 Emerging Technologies Building, 3120 TAMU, College Station, TX 77843 USA
| | - Maygan K. McFarlin
- Department of Biomedical Engineering, Texas A&M University, 5016 Emerging Technologies Building, 3120 TAMU, College Station, TX 77843 USA
| | - Claudia A. Collier
- Department of Biomedical Engineering, Texas A&M University, 5016 Emerging Technologies Building, 3120 TAMU, College Station, TX 77843 USA
| | - Vibha S. Sheshadri
- Department of Biomedical Engineering, Texas A&M University, 5016 Emerging Technologies Building, 3120 TAMU, College Station, TX 77843 USA
| | - Anirudh K. Madyastha
- Department of Biomedical Engineering, Texas A&M University, 5016 Emerging Technologies Building, 3120 TAMU, College Station, TX 77843 USA
| | - John Paul M. Gorley
- Department of Biomedical Engineering, Texas A&M University, 5016 Emerging Technologies Building, 3120 TAMU, College Station, TX 77843 USA
| | - Spencer C. Solberg
- Department of Biomedical Engineering, Texas A&M University, 5016 Emerging Technologies Building, 3120 TAMU, College Station, TX 77843 USA
| | - Amber N. Stratman
- Department of Cell Biology and Physiology, Washington University School of Medicine in St. Louis, St. Louis, MO USA
| | - Shreya A. Raghavan
- Department of Biomedical Engineering, Texas A&M University, 5016 Emerging Technologies Building, 3120 TAMU, College Station, TX 77843 USA
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX USA
| |
Collapse
|
21
|
Lučić I, Kurtović M, Mlinarić M, Piteša N, Čipak Gašparović A, Sabol M, Milković L. Deciphering Common Traits of Breast and Ovarian Cancer Stem Cells and Possible Therapeutic Approaches. Int J Mol Sci 2023; 24:10683. [PMID: 37445860 DOI: 10.3390/ijms241310683] [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: 05/06/2023] [Revised: 06/21/2023] [Accepted: 06/23/2023] [Indexed: 07/15/2023] Open
Abstract
Breast cancer (BC) and ovarian cancer (OC) are among the most common and deadly cancers affecting women worldwide. Both are complex diseases with marked heterogeneity. Despite the induction of screening programs that increase the frequency of earlier diagnosis of BC, at a stage when the cancer is more likely to respond to therapy, which does not exist for OC, more than 50% of both cancers are diagnosed at an advanced stage. Initial therapy can put the cancer into remission. However, recurrences occur frequently in both BC and OC, which are highly cancer-subtype dependent. Therapy resistance is mainly attributed to a rare subpopulation of cells, named cancer stem cells (CSC) or tumor-initiating cells, as they are capable of self-renewal, tumor initiation, and regrowth of tumor bulk. In this review, we will discuss the distinctive markers and signaling pathways that characterize CSC, their interactions with the tumor microenvironment, and the strategies they employ to evade immune surveillance. Our focus will be on identifying the common features of breast cancer stem cells (BCSC) and ovarian cancer stem cells (OCSC) and suggesting potential therapeutic approaches.
Collapse
Affiliation(s)
- Ivan Lučić
- Laboratory for Oxidative Stress, Division of Molecular Medicine, Ruđer Bošković Institute, 10000 Zagreb, Croatia
| | - Matea Kurtović
- Laboratory for Hereditary Cancer, Division of Molecular Medicine, Ruđer Bošković Institute, 10000 Zagreb, Croatia
| | - Monika Mlinarić
- Laboratory for Oxidative Stress, Division of Molecular Medicine, Ruđer Bošković Institute, 10000 Zagreb, Croatia
| | - Nikolina Piteša
- Laboratory for Hereditary Cancer, Division of Molecular Medicine, Ruđer Bošković Institute, 10000 Zagreb, Croatia
| | - Ana Čipak Gašparović
- Laboratory for Oxidative Stress, Division of Molecular Medicine, Ruđer Bošković Institute, 10000 Zagreb, Croatia
| | - Maja Sabol
- Laboratory for Hereditary Cancer, Division of Molecular Medicine, Ruđer Bošković Institute, 10000 Zagreb, Croatia
| | - Lidija Milković
- Laboratory for Oxidative Stress, Division of Molecular Medicine, Ruđer Bošković Institute, 10000 Zagreb, Croatia
| |
Collapse
|
22
|
Wu MC, Yu HW, Chen YQ, Ou MH, Serrano R, Huang GL, Wang YK, Lin KH, Fan YJ, Wu CC, Del Álamo JC, Chiou A, Chien S, Kuo JC. Early committed polarization of intracellular tension in response to cell shape determines the osteogenic differentiation of mesenchymal stromal cells. Acta Biomater 2023; 163:287-301. [PMID: 36328121 PMCID: PMC11389728 DOI: 10.1016/j.actbio.2022.10.052] [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: 04/15/2022] [Revised: 10/24/2022] [Accepted: 10/25/2022] [Indexed: 11/07/2022]
Abstract
Within the heterogeneous tissue architecture, a comprehensive understanding of how cell shapes regulate cytoskeletal mechanics by adjusting focal adhesions (FAs) signals to correlate with the lineage commitment of mesenchymal stromal cells (MSCs) remains obscure. Here, via engineered extracellular matrices, we observed that the development of mature FAs, coupled with a symmetrical pattern of radial fiber bundles, appeared at the right-angle vertices in cells with square shape. While circular cells aligned the transverse fibers parallel to the cell edge, and moved them centripetally in a counter-clockwise direction, symmetrical bundles of radial fibers at the vertices of square cells disrupted the counter-clockwise swirling and bridged the transverse fibers to move centripetally. In square cells, the contractile force, generated by the myosin IIA-enriched transverse fibers, were concentrated and transmitted outwards along the symmetrical bundles of radial fibers, to the extracellular matrix through FAs, and thereby driving FA organization and maturation. The symmetrical radial fiber bundles concentrated the transverse fibers contractility inward to the linkage between the actin cytoskeleton and the nuclear envelope. The tauter cytoskeletal network adjusted the nuclear-actomyosin force balance to cause nuclear deformability and to increase nuclear translocation of the transcription co-activator YAP, which in turn modulated the switch in MSC commitment. Thus, FAs dynamically respond to geometric cues and remodel actin cytoskeletal network to re-distribute intracelluar tension towards the cell nucleus, and thereby controlling YAP mechanotransduction signaling in regulating MSC fate decision. STATEMENT OF SIGNIFICANCE: We decipher how cellular mechanics is self-organized depending on extracellular geometric features to correlate with mesenchymal stromal cell lineage commitment. In response to geometry constrains on cell morphology, symmetrical radial fiber bundles are assembled and clustered depending on the maturation state of focal adhesions and bridge with the transverse fibers, and thereby establishing the dynamic cytoskeletal network. Contractile force, generated by the myosin-IIA-enriched transverse fibers, is transmitted and dynamically drives the retrograde movement of the actin cytoskeletal network, which appropriately adjusts the nuclear-actomyosin force balance and deforms the cell nucleus for YAP mechano-transduction signaling in regulating mesenchymal stromal cell fate decision.
Collapse
Affiliation(s)
- Ming-Chung Wu
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Helen Wenshin Yu
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan; Institute of Biophotonics, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Yin-Quan Chen
- Cancer Progression Research Center, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Meng-Hsin Ou
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Ricardo Serrano
- Department of Bioengineering and Institute of Engineering in Medicine, University of California at San Diego, La Jolla, CA 92093, USA; Department of Mechanical and Aerospace Engineering, University of California at San Diego, La Jolla, CA 92093, USA; Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Guan-Lin Huang
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Yang-Kao Wang
- Department of Cell Biology and Anatomy, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Kung-Hui Lin
- Institute of Physics, Academia Sinica, Taipei 11529, Taiwan
| | - Yu-Jui Fan
- School of Biomedical Engineering, Taipei Medical University, Taipei 110, Taiwan
| | - Chi-Chang Wu
- Department of Electronic Engineering, National Chin-Yi University of Technology, Taichung 411030, Taiwan
| | - Juan C Del Álamo
- Department of Bioengineering and Institute of Engineering in Medicine, University of California at San Diego, La Jolla, CA 92093, USA; Department of Mechanical and Aerospace Engineering, University of California at San Diego, La Jolla, CA 92093, USA; Center for Cardiovascular Biology, University of Washington, School of Medicine, Seattle, WA, 98109, USA; Mechanical Engineering Department, University of Washington, Seattle, WA, 98195, USA
| | - Arthur Chiou
- Institute of Biophotonics, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Shu Chien
- Department of Bioengineering and Institute of Engineering in Medicine, University of California at San Diego, La Jolla, CA 92093, USA
| | - Jean-Cheng Kuo
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan; Cancer Progression Research Center, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan.
| |
Collapse
|
23
|
Li Z, Zheng C, Jiang P, Xu X, Tang Y, Dou L. Human digested dentin matrix for dentin regeneration and the applicative potential in vital pulp therapy. J Endod 2023:S0099-2399(23)00238-8. [PMID: 37150293 DOI: 10.1016/j.joen.2023.04.010] [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: 01/12/2023] [Revised: 04/22/2023] [Accepted: 04/23/2023] [Indexed: 05/09/2023]
Abstract
INTRODUCTION Human dentin is a natural acellular matrix with excellent reported biocompatibility. The aim was to fabricate a novel dentin matrix material from human dentin and investigate its applicative potential for vital pulp therapy. METHODS Digested dentin matrix extract (DDME) was fabricated using controlled enzymatic digestion under acidic conditions. The surfaces and biocompatibility of DDME were then investigated, with its effects on the odontogenic differentiation of human dental pulp cells (hDPCs) also studied. The ability of DDME to induce mineralization was assessed in a nude mouse model. The performance of DDME as a pulp capping agent was evaluated in an in-situ rat model. The molecular mechanism was verified by mRNA sequencing. RESULTS A novel type of dentin matrix material with a uniform size of 8 μm was fabricated. DDME had a similar band compared with grinded dentin matrix, with a smaller size and more uneven surface, as detected by Fourier Transform Infrared Spectrometer (FTIR) and X-ray photoelectron spectroscopy (XPS). DDME at low concentrations did not affect hDPCs viability or proliferation, but enhanced runt-related transcription factor 2, dentin matrix acidic phosphoprotein 1 and collagen 1A1 expression in hDPCs in vitro. DDME was superior to HA-TCP in dentin-like mineralized tissue formation after subcutaneous transplantation. In the rat model of pulpotomy, DDME showed visible curative effects. The underlying mechanism may be the inhibition of Hippo signaling following DDME treatment. DDME promoted Yes-associated protein (YAP) 1 nuclear influx, thereby enhancing the expression of DMP-1, which was reversed by YAP inhibitor treatment. CONCLUSIONS Human DDME can be used as a biomaterial for dentin regeneration. The combined application of DDME and current pulp capping agents is a potential choice for vital pulp therapy.
Collapse
Affiliation(s)
- Zheng Li
- 6 Stomatological Hospital of Chongqing Medical University, 426#, Song Shi Bei Road, Chongqing, 401147, China; Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Medical, University, Chongqing, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing Medical University, Chongqing, China
| | - Chengxiang Zheng
- 6 Stomatological Hospital of Chongqing Medical University, 426#, Song Shi Bei Road, Chongqing, 401147, China; Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Medical, University, Chongqing, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing Medical University, Chongqing, China
| | - Peiru Jiang
- 6 Stomatological Hospital of Chongqing Medical University, 426#, Song Shi Bei Road, Chongqing, 401147, China; Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Medical, University, Chongqing, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing Medical University, Chongqing, China
| | - Xiaoqi Xu
- 6 Stomatological Hospital of Chongqing Medical University, 426#, Song Shi Bei Road, Chongqing, 401147, China; Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Medical, University, Chongqing, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing Medical University, Chongqing, China
| | - Yin Tang
- University of Southern California Herman Ostrow School of Dentistry, Los Angeles, CA, USA; School of Dental Medicine, Western University of Medical Sciences, Pomona, CA, USA
| | - Lei Dou
- 6 Stomatological Hospital of Chongqing Medical University, 426#, Song Shi Bei Road, Chongqing, 401147, China; Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Medical, University, Chongqing, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
24
|
Huang HC, Wang TY, Rousseau J, Mungaray M, Michaud C, Plaisier C, Chen ZB, Wang KC. Lesion-specific suppression of YAP/TAZ by biomimetic nanodrug ameliorates atherosclerosis development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.24.537992. [PMID: 37163067 PMCID: PMC10168204 DOI: 10.1101/2023.04.24.537992] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Atherosclerosis, characterized by the buildup of lipid-rich plaque on the vessel wall, is the primary cause of myocardial infarction and ischemic stroke. Recent studies have demonstrated that dysregulation of yes-associated protein 1 (YAP) and transcriptional coactivator with PDZ-binding domain (TAZ) contributes to plaque development, making YAP/TAZ potential therapeutic targets. However, systemic modulation of YAP/TAZ expression or activities risks serious off-target effects, limiting clinical applicability. To address the challenge, this study develops monocyte membrane-coated nanoparticles (MoNP) as a drug delivery vehicle targeting activated endothelium lining the plaque surface and utilizes MoNP to deliver verteporfin (VP), a potent YAP/TAZ inhibitor, for lesion-specific treatment of atherosclerosis. The results reveal that MoNP significantly enhance payload delivery to inflamed endothelial cells (EC) while avoiding phagocytic cells, and preferentially accumulate in atherosclerotic regions. MoNP-mediated delivery of VP substantially reduces YAP/TAZ expression, suppressing inflammatory gene expression and macrophage infiltration in cultured EC and mouse arteries exposed to atherogenic stimuli. Importantly, this lesion-targeted VP nanodrug effectively decreases plaque development in mice without causing noticeable histopathological changes in major organs. Collectively, these findings demonstrate a plaque-targeted and pathway-specific biomimetic nanodrug, potentially leading to safer and more effective treatments for atherosclerosis.
Collapse
|
25
|
Yang Q, Al-Hendy A. Update on the Role and Regulatory Mechanism of Extracellular Matrix in the Pathogenesis of Uterine Fibroids. Int J Mol Sci 2023; 24:5778. [PMID: 36982852 PMCID: PMC10051203 DOI: 10.3390/ijms24065778] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 02/22/2023] [Accepted: 03/08/2023] [Indexed: 03/30/2023] Open
Abstract
Uterine fibroids (UFs), also known as leiomyomas, are benign tumors of the myometrium affecting over 70% of women worldwide, particularly women of color. Although benign, UFs are associated with significant morbidity; they are the primary indication for hysterectomy and a major source of gynecologic and reproductive dysfunction, ranging from menorrhagia and pelvic pain to infertility, recurrent miscarriage, and preterm labor. So far, the molecular mechanisms underlying the pathogenesis of UFs are still quite limited. A knowledge gap needs to be filled to help develop novel strategies that will ultimately facilitate the development of therapies and improve UF patient outcomes. Excessive ECM accumulation and aberrant remodeling are crucial for fibrotic diseases and excessive ECM deposition is the central characteristics of UFs. This review summarizes the recent progress of ascertaining the biological functions and regulatory mechanisms in UFs, from the perspective of factors regulating ECM production, ECM-mediated signaling, and pharmacological drugs targeting ECM accumulation. In addition, we provide the current state of knowledge by discussing the molecular mechanisms underlying the regulation and emerging role of the extracellular matrix in the pathogenesis of UFs and in applications. Comprehensive and deeper insights into ECM-mediated alterations and interactions in cellular events will help develop novel strategies to treat patients with this common tumor.
Collapse
Affiliation(s)
- Qiwei Yang
- Department of Obstetrics and Gynecology, University of Chicago, Chicago, IL 60637, USA;
| | | |
Collapse
|
26
|
Ma H, Tian T, Cui Z. Targeting ovarian cancer stem cells: a new way out. Stem Cell Res Ther 2023; 14:28. [PMID: 36788591 PMCID: PMC9926632 DOI: 10.1186/s13287-023-03244-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 01/18/2023] [Indexed: 02/16/2023] Open
Abstract
Ovarian cancer (OC) is the most lethal gynecological malignancy due to tumor heterogeneity, the lack of reliable early diagnosis methods and the high incidence of chemoresistant recurrent disease. Although there are developments in chemotherapies and surgical techniques to improve the overall survival of OC patients, the 5-year survival of advanced OC patients is still low. To improve the prognosis of OC patients, it is important to search for novel therapeutic approaches. Cancer stem cells (CSCs) are a subpopulation of tumor cells that participate in tumor growth, metastasis and chemoresistance. It is important to study the role of CSCs in a highly heterogeneous disease such as OC, which may be significant to a better understanding of the oncogenetic and metastatic pathways of the disease and to develop novel strategies against its progression and platinum resistance. Here, we summarized the current findings about targeting methods against ovarian cancer stem cells, including related signaling pathways, markers and drugs, to better manage OC patients using CSC-based therapeutic strategies.
Collapse
Affiliation(s)
- Huiying Ma
- grid.412521.10000 0004 1769 1119Department of Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Tian Tian
- grid.412521.10000 0004 1769 1119Department of Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, People’s Republic of China
| | - Zhumei Cui
- Department of Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China.
| |
Collapse
|
27
|
Genetic Alterations and Deregulation of Hippo Pathway as a Pathogenetic Mechanism in Bone and Soft Tissue Sarcoma. Cancers (Basel) 2022; 14:cancers14246211. [PMID: 36551696 PMCID: PMC9776600 DOI: 10.3390/cancers14246211] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/13/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
The Hippo pathway is an evolutionarily conserved modulator of developmental biology with a key role in tissue and organ size regulation under homeostatic conditions. Like other signaling pathways with a significant role in embryonic development, the deregulation of Hippo signaling contributes to oncogenesis. Central to the Hippo pathway is a conserved cascade of adaptor proteins and inhibitory kinases that converge and regulate the activity of the oncoproteins YAP and TAZ, the final transducers of the pathway. Elevated levels and aberrant activation of YAP and TAZ have been described in many cancers. Though most of the studies describe their pervasive activation in epithelial neoplasms, there is increasing evidence pointing out its relevance in mesenchymal malignancies as well. Interestingly, somatic or germline mutations in genes of the Hippo pathway are scarce compared to other signaling pathways that are frequently disrupted in cancer. However, in the case of sarcomas, several examples of genetic alteration of Hippo members, including gene fusions, have been described during the last few years. Here, we review the current knowledge of Hippo pathway implication in sarcoma, describing mechanistic hints recently reported in specific histological entities and how these alterations represent an opportunity for targeted therapy in this heterogeneous group of neoplasm.
Collapse
|
28
|
Carigga Gutierrez NM, Pujol-Solé N, Arifi Q, Coll JL, le Clainche T, Broekgaarden M. Increasing cancer permeability by photodynamic priming: from microenvironment to mechanotransduction signaling. Cancer Metastasis Rev 2022; 41:899-934. [PMID: 36155874 DOI: 10.1007/s10555-022-10064-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 09/06/2022] [Indexed: 01/25/2023]
Abstract
The dense cancer microenvironment is a significant barrier that limits the penetration of anticancer agents, thereby restraining the efficacy of molecular and nanoscale cancer therapeutics. Developing new strategies to enhance the permeability of cancer tissues is of major interest to overcome treatment resistance. Nonetheless, early strategies based on small molecule inhibitors or matrix-degrading enzymes have led to disappointing clinical outcomes by causing increased chemotherapy toxicity and promoting disease progression. In recent years, photodynamic therapy (PDT) has emerged as a novel approach to increase the permeability of cancer tissues. By producing excessive amounts of reactive oxygen species selectively in the cancer microenvironment, PDT increases the accumulation, penetration depth, and efficacy of chemotherapeutics. Importantly, the increased cancer permeability has not been associated to increased metastasis formation. In this review, we provide novel insights into the mechanisms by which this effect, called photodynamic priming, can increase cancer permeability without promoting cell migration and dissemination. This review demonstrates that PDT oxidizes and degrades extracellular matrix proteins, reduces the capacity of cancer cells to adhere to the altered matrix, and interferes with mechanotransduction pathways that promote cancer cell migration and differentiation. Significant knowledge gaps are identified regarding the involvement of critical signaling pathways, and to which extent these events are influenced by the complicated PDT dosimetry. Addressing these knowledge gaps will be vital to further develop PDT as an adjuvant approach to improve cancer permeability, demonstrate the safety and efficacy of this priming approach, and render more cancer patients eligible to receive life-extending treatments.
Collapse
Affiliation(s)
| | - Núria Pujol-Solé
- Université Grenoble Alpes, Inserm U 1209, CNRS UMR 5309, Institute for Advanced Biosciences, 38000, Grenoble, France
| | - Qendresa Arifi
- Université Grenoble Alpes, Inserm U 1209, CNRS UMR 5309, Institute for Advanced Biosciences, 38000, Grenoble, France
| | - Jean-Luc Coll
- Université Grenoble Alpes, Inserm U 1209, CNRS UMR 5309, Institute for Advanced Biosciences, 38000, Grenoble, France
| | - Tristan le Clainche
- Université Grenoble Alpes, Inserm U 1209, CNRS UMR 5309, Institute for Advanced Biosciences, 38000, Grenoble, France.
| | - Mans Broekgaarden
- Université Grenoble Alpes, Inserm U 1209, CNRS UMR 5309, Institute for Advanced Biosciences, 38000, Grenoble, France.
| |
Collapse
|
29
|
FSH Regulates YAP-TEAD Transcriptional Activity in Bovine Granulosa Cells to Allow the Future Dominant Follicle to Exert Its Augmented Estrogenic Capacity. Int J Mol Sci 2022; 23:ijms232214160. [PMID: 36430640 PMCID: PMC9693326 DOI: 10.3390/ijms232214160] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 11/12/2022] [Accepted: 11/14/2022] [Indexed: 11/18/2022] Open
Abstract
The molecular mechanisms that drive the granulosa cells' (GC) differentiation into a more estrogenic phenotype during follicular divergence and establishment of follicle dominance have not been completely elucidated. The main Hippo signaling effector, YAP, has, however, emerged as a potential key player to explain such complex processes. Studies using rat and bovine GC demonstrate that, in conditions where the expression of the classic YAP-TEAD target gene tissue growth factor (CTGF) is augmented, CYP19A1 expression and activity and, consequently, estradiol (E2) secretion are reduced. These findings led us to hypothesize that, during ovarian follicular divergence in cattle, FSH downregulates YAP-TEAD-dependent transcriptional activity in GC to allow the future dominant follicle to exert its augmented estrogenic capacity. To address this, we performed a series of experiments employing distinct bovine models. Our in vitro and ex vivo experiments indicated that indeed FSH downregulates, in a concentration-dependent manner, mRNA levels not only for CTGF but also for the other classic YAP-TEAD transcriptional target genes ANKRD1 and CYR61 by a mechanism that involves increased YAP phosphorylation. To better elucidate the functional importance of such FSH-induced YAP activity regulation, we then cultured GC in the presence of verteporfin (VP) or peptide 17 (P17), two pharmacological inhibitors known to interfere with YAP binding to TEADs. The results showed that both VP and P17 increased CYP19A1 basal mRNA levels in a concentration-dependent manner. Most interestingly, by using GC samples obtained in vivo from dominant vs. subordinate follicles, we found that mRNA levels for CTGF, CYR61, and ANKRD1 are higher in subordinate follicles following the follicular divergence. Taken together, our novel results demonstrate that YAP transcriptional activity is regulated in bovine granulosa cells to allow the increased estrogenic capacity of the selected dominant follicle.
Collapse
|
30
|
van IJzendoorn DG, Matusiak M, Charville GW, Spierenburg G, Varma S, Colburg DR, van de Sande MA, van Langevelde K, Mohler DG, Ganjoo KN, Bui NQ, Avedian RS, Bovée JV, Steffner R, West RB, van de Rijn M. Interactions in CSF1-Driven Tenosynovial Giant Cell Tumors. Clin Cancer Res 2022; 28:4934-4946. [PMID: 36007098 PMCID: PMC9660542 DOI: 10.1158/1078-0432.ccr-22-1898] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/25/2022] [Accepted: 08/23/2022] [Indexed: 01/24/2023]
Abstract
PURPOSE A major component of cells in tenosynovial giant cell tumor (TGCT) consists of bystander macrophages responding to CSF1 that is overproduced by a small number of neoplastic cells with a chromosomal translocation involving the CSF1 gene. An autocrine loop was postulated where the neoplastic cells would be stimulated through CSF1R expressed on their surface. Here, we use single-cell RNA sequencing (scRNA-seq) to investigate cellular interactions in TGCT. EXPERIMENTAL DESIGN A total of 18,788 single cells from three TGCT and two giant cell tumor of bone (GCTB) samples underwent scRNA-seq. The three TGCTs were additionally analyzed using long-read RNA sequencing. Immunofluorescence and IHC for a range of markers were used to validate and extend the scRNA-seq findings. RESULTS Two recurrent neoplastic cell populations were identified in TGCT that are highly similar to nonneoplastic synoviocytes. We identified GFPT2 as a marker that highlights the neoplastic cells in TCGT. We show that the neoplastic cells themselves do not express CSF1R. We identified overlapping MAB features between the giant cells in TGCT and GCTB. CONCLUSIONS The neoplastic cells in TGCT are highly similar to nonneoplastic synoviocytes. The lack of CSF1R on the neoplastic cells indicates they may be unaffected by current therapies. High expression of GFPT2 in the neoplastic cells is associated with activation of the YAP1/TAZ pathway. In addition, we identified expression of the platelet-derived growth factor receptor in the neoplastic cells. These findings suggest two additional pathways to target in this tumor.
Collapse
Affiliation(s)
| | - Magdalena Matusiak
- Department of Pathology, Stanford University School of Medicine, Stanford, California
| | - Gregory W. Charville
- Department of Pathology, Stanford University School of Medicine, Stanford, California
| | - Geert Spierenburg
- Department of Orthopedic Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | - Sushama Varma
- Department of Pathology, Stanford University School of Medicine, Stanford, California
| | - Deana R.C. Colburg
- Department of Pathology, Stanford University School of Medicine, Stanford, California
| | | | | | - David G. Mohler
- Department of Orthopedic Surgery, Stanford University, Stanford, California
| | - Kristen N. Ganjoo
- Division of Oncology, Department of Medicine, Stanford University, Stanford, California
| | - Nam Q. Bui
- Division of Oncology, Department of Medicine, Stanford University, Stanford, California
| | - Raffi S. Avedian
- Department of Orthopedic Surgery, Stanford University, Stanford, California
| | - Judith V.M.G. Bovée
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands
| | - Robert Steffner
- Department of Orthopedic Surgery, Stanford University, Stanford, California
| | - Robert B. West
- Department of Pathology, Stanford University School of Medicine, Stanford, California
| | - Matt van de Rijn
- Department of Pathology, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
31
|
Ebrahimighaei R, Sala-Newby GB, Hudson C, Kimura TE, Hathway T, Hawkins J, McNeill MC, Richardson R, Newby AC, Bond M. Combined role for YAP-TEAD and YAP-RUNX2 signalling in substrate-stiffness regulation of cardiac fibroblast proliferation. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2022; 1869:119329. [PMID: 35905788 PMCID: PMC7616274 DOI: 10.1016/j.bbamcr.2022.119329] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 07/16/2022] [Accepted: 07/20/2022] [Indexed: 06/15/2023]
Abstract
Cardiac fibrosis is associated with increased stiffness of the myocardial extracellular matrix (ECM) in part mediated by increased cardiac fibroblast proliferation However, our understanding of the mechanisms regulating cardiac fibroblast proliferation are incomplete. Here we characterise a novel mechanism involving a combined activation of Yes-associated protein (YAP) targets RUNX Family Transcription Factor 2 (RUNX2) and TEA Domain Transcription Factor (TEAD). We demonstrate that cardiac fibroblast proliferation is enhanced by interaction with a stiff ECM compared to a soft ECM. This is associated with activation of the transcriptional co-factor, YAP. We demonstrate that this stiffness induced activation of YAP enhances the transcriptional activity of both TEAD and RUNX2 transcription factors. Inhibition of either TEAD or RUNX2, using gene silencing, expression of dominant-negative mutants or pharmacological inhibition, reduces cardiac fibroblast proliferation. Using mutants of YAP, defective in TEAD or RUNX2 activation ability, we demonstrate a dual role of YAP-mediated activation of TEAD and RUNX2 for substrate stiffness induced cardiac fibroblast proliferation. Our data highlights a previously unrecognised role of YAP mediated RUNX2 activation for cardiac fibroblast proliferation in response to increased ECM stiffness.
Collapse
Affiliation(s)
- Reza Ebrahimighaei
- School of Translational Health Sciences, Faculty of Health Sciences, University of Bristol, Research Floor Level 7, Bristol Royal Infirmary, Bristol BS2 8HW, UK
| | - Graciela B Sala-Newby
- School of Translational Health Sciences, Faculty of Health Sciences, University of Bristol, Research Floor Level 7, Bristol Royal Infirmary, Bristol BS2 8HW, UK
| | - Claire Hudson
- School of Translational Health Sciences, Faculty of Health Sciences, University of Bristol, Research Floor Level 7, Bristol Royal Infirmary, Bristol BS2 8HW, UK
| | - Tomomi E Kimura
- School of Translational Health Sciences, Faculty of Health Sciences, University of Bristol, Research Floor Level 7, Bristol Royal Infirmary, Bristol BS2 8HW, UK
| | - Tom Hathway
- School of Translational Health Sciences, Faculty of Health Sciences, University of Bristol, Research Floor Level 7, Bristol Royal Infirmary, Bristol BS2 8HW, UK
| | - Joseph Hawkins
- School of Translational Health Sciences, Faculty of Health Sciences, University of Bristol, Research Floor Level 7, Bristol Royal Infirmary, Bristol BS2 8HW, UK
| | - Madeleine C McNeill
- School of Translational Health Sciences, Faculty of Health Sciences, University of Bristol, Research Floor Level 7, Bristol Royal Infirmary, Bristol BS2 8HW, UK
| | - Rebecca Richardson
- School of Translational Health Sciences, Faculty of Health Sciences, University of Bristol, Research Floor Level 7, Bristol Royal Infirmary, Bristol BS2 8HW, UK
| | - Andrew C Newby
- School of Translational Health Sciences, Faculty of Health Sciences, University of Bristol, Research Floor Level 7, Bristol Royal Infirmary, Bristol BS2 8HW, UK
| | - Mark Bond
- School of Translational Health Sciences, Faculty of Health Sciences, University of Bristol, Research Floor Level 7, Bristol Royal Infirmary, Bristol BS2 8HW, UK.
| |
Collapse
|
32
|
Human embryonic stem cell-specific role of YAP in maintenance of self-renewal and survival. Cell Mol Life Sci 2022; 79:544. [PMID: 36219276 DOI: 10.1007/s00018-022-04558-x] [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: 05/24/2022] [Revised: 08/30/2022] [Accepted: 09/06/2022] [Indexed: 11/03/2022]
Abstract
Human embryonic stem cells (hESCs) have unique characteristics, such as self-renewal and pluripotency, which are distinct from those of other cell types. These characteristics of hESCs are tightly regulated by complex signaling mechanisms. In this study, we demonstrate that yes-associated protein (YAP) functions in an hESC-specific manner to maintain self-renewal and survival in hESCs. hESCs were highly sensitive to YAP downregulation to promote cell survival. Interestingly, hESCs displayed dynamic changes in YAP expression in response to YAP downregulation. YAP was critical for the maintenance of self-renewal. Additionally, the function of YAP in maintenance of self-renewal and cell survival was hESC-specific. Doxycycline upregulated YAP in hESCs and attenuated the decreased cell survival induced by YAP downregulation. However, decreased expression of self-renewal markers triggered by YAP downregulation and neural/cardiac differentiation were affected by doxycycline treatment. Collectively, the results reveal the mechanism underlying the role of YAP and the novel function of doxycycline in hESCs.
Collapse
|
33
|
Fu J, McGrath NA, Lee J, Wang X, Brar G, Xie C. Verteporfin synergizes the efficacy of anti-PD-1 in cholangiocarcinoma. Hepatobiliary Pancreat Dis Int 2022; 21:485-492. [PMID: 35307294 PMCID: PMC9463402 DOI: 10.1016/j.hbpd.2022.03.006] [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: 07/31/2021] [Accepted: 03/01/2022] [Indexed: 02/05/2023]
Abstract
BACKGROUND Cholangiocarcinoma (CCA) is one of the primary hepatobiliary malignant neoplasms with only 10% of 5-year survival rate. Promising immunotherapy with the blockade of immune checkpoints has no clear benefit in CCA. The inhibition of YAP1 signaling by verteporfin has shown encouraging results by inhibiting cell proliferation and inducing apoptosis. This study aimed to evaluate the potential benefit of the combination of verteporfin and anti-programmed cell death 1 (PD-1) in CCA mouse model. METHODS We assessed the cytotoxicity of verteporfin in human CCA cell lines in vitro, including both intrahepatic CCA and extrahepatic CCA cells. We examined the in vitro effect of verteporfin on cell proliferation, apoptosis, and stemness. We evaluated the in vivo efficacy of verteporfin, anti-PD-1, and a combination of both in subcutaneous CCA mouse model. RESULTS Our study showed that verteporfin reduced tumor cell growth and enhanced apoptosis of human CCA tumor cells in vitro in a dose-dependent fashion. Nevertheless, verteporfin impaired stemness evidenced by reduced spheroid formation and colony formation, decreased numbers of cells with aldehyde dehydrogenase activity and positive cancer stem cell markers (all P < 0.05). The combination of verteporfin and anti-PD-1 reduced tumor burden in CCA subcutaneous SB1 tumor model compared to either agent alone. CONCLUSIONS Verteporfin exhibits antitumor effects in both intrahepatic and extrahepatic CCA cell lines and the combination with anti-PD-1 inhibited tumor growth.
Collapse
Affiliation(s)
- Jianyang Fu
- Thoracic and Gastrointestinal Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Nicole A McGrath
- Thoracic and Gastrointestinal Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jihye Lee
- Thoracic and Gastrointestinal Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Xin Wang
- Thoracic and Gastrointestinal Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Gagandeep Brar
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10065, USA
| | - Changqing Xie
- Thoracic and Gastrointestinal Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
34
|
Capicua suppresses YAP1 to limit tumorigenesis and maintain drug sensitivity in human cancer. Cell Rep 2022; 41:111443. [DOI: 10.1016/j.celrep.2022.111443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 07/28/2022] [Accepted: 09/09/2022] [Indexed: 11/23/2022] Open
|
35
|
Fowler EW, van Venrooy EJ, Witt RL, Jia X. A TGFβR inhibitor represses keratin-7 expression in 3D cultures of human salivary gland progenitor cells. Sci Rep 2022; 12:15008. [PMID: 36056161 PMCID: PMC9440137 DOI: 10.1038/s41598-022-19253-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 08/26/2022] [Indexed: 11/09/2022] Open
Abstract
Salivary gland tissue engineering offers an attractive alternative for the treatment of radiation-induced xerostomia. Key to the success of this approach is the maintenance and expansion of secretory acinar cells in vitro. However, recent studies revealed that in vitro culture of primary salivary gland epithelial cells led to undesirable upregulation of the expression of keratin-7 (K7), a marker of ductal phenotype and frequently associated with cellular stress. We have previously shown that hyaluronic acid (HA)-based, RGDSP-decorated hydrogels support the 3D growth and assembly of primary human salivary gland stem/progenitor cells (hS/PCs). Here, we investigate whether the RGDSP culture also promotes K7 expression, and if so, what factors govern the K7 expression. Compared to hS/PCs maintained in blank HA gels, those grown in RGDSP cultures expressed a significantly higher level of K7. In other tissues, various transforming growth factor-β (TGF-β) superfamily members are reported to regulate K7 expression. Similarly, our immunoblot array and ELISA experiments confirmed the increased expression of TGF-β1 and growth/differentiation factor-15 (GDF-15) in RGDSP cultures. However, 2D model studies show that only TGF-β1 is required to induce K7 expression in hS/PCs. Immunocytochemical analysis of the intracellular effectors of TGF-β signaling, SMAD 2/3, further confirmed the elevated TGF-β signaling in RGDSP cultures. To maximize the regenerative potential of h/SPCs, cultures were treated with a pharmacological inhibitor of TGF-β receptor, A83-01. Our results show that A83-01 treatment can repress K7 expression not only in 3D RGDSP cultures but also under 2D conditions with exogenous TGF-β1. Collectively, we provide a link between TGF-β signaling and K7 expression in hS/PC cultures and demonstrate the effectiveness of TGF-β inhibition to repress K7 expression while maintaining the ability of RGDSP-conjugated HA gels to facilitate the rapid development of amylase expressing spheroids. These findings represent an important step towards regenerating salivary function with a tissue-engineered salivary gland.
Collapse
Affiliation(s)
- Eric W Fowler
- Department of Materials Science and Engineering, University of Delaware, Newark, DE, 19716, USA.
| | - Emmett J van Venrooy
- Department of Biological Sciences, University of Delaware, Newark, DE, 19716, USA
| | - Robert L Witt
- Helen F. Graham Cancer Center and Research Institute, Christiana Care, Newark, DE, 19713, USA
| | - Xinqiao Jia
- Department of Materials Science and Engineering, University of Delaware, Newark, DE, 19716, USA.
- Department of Biological Sciences, University of Delaware, Newark, DE, 19716, USA.
- Department of Biomedical Engineering, University of Delaware, Newark, DE, 19716, USA.
- Delaware Biotechnology Institute, 590 Avenue 1743, Newark, DE, 19713, USA.
| |
Collapse
|
36
|
Shi J, Xu J, Li Y, Li B, Ming H, Nice EC, Huang C, Li Q, Wang C. Drug repurposing in cancer neuroscience: From the viewpoint of the autophagy-mediated innervated niche. Front Pharmacol 2022; 13:990665. [PMID: 36105204 PMCID: PMC9464986 DOI: 10.3389/fphar.2022.990665] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 08/01/2022] [Indexed: 11/13/2022] Open
Abstract
Based on the bidirectional interactions between neurology and cancer science, the burgeoning field “cancer neuroscience” has been proposed. An important node in the communications between nerves and cancer is the innervated niche, which has physical contact with the cancer parenchyma or nerve located in the proximity of the tumor. In the innervated niche, autophagy has recently been reported to be a double-edged sword that plays a significant role in maintaining homeostasis. Therefore, regulating the innervated niche by targeting the autophagy pathway may represent a novel therapeutic strategy for cancer treatment. Drug repurposing has received considerable attention for its advantages in cost-effectiveness and safety. The utilization of existing drugs that potentially regulate the innervated niche via the autophagy pathway is therefore a promising pharmacological approach for clinical practice and treatment selection in cancer neuroscience. Herein, we present the cancer neuroscience landscape with an emphasis on the crosstalk between the innervated niche and autophagy, while also summarizing the underlying mechanisms of candidate drugs in modulating the autophagy pathway. This review provides a strong rationale for drug repurposing in cancer treatment from the viewpoint of the autophagy-mediated innervated niche.
Collapse
Affiliation(s)
- Jiayan Shi
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Jia Xu
- Department of Pharmacology, Provincial Key Laboratory of Pathophysiology, Ningbo University School of Medicine, Ningbo, China
| | - Yang Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Bowen Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Hui Ming
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Edouard C. Nice
- Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, Australia
| | - Canhua Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Qifu Li
- Department of Neurology and Key Laboratory of Brain Science Research and Transformation in Tropical Environment of Hainan Province, The First Affiliated Hospital, Hainan Medical University, Haikou, China
- *Correspondence: Qifu Li, ; Chuang Wang,
| | - Chuang Wang
- Department of Pharmacology, Provincial Key Laboratory of Pathophysiology, Ningbo University School of Medicine, Ningbo, China
- *Correspondence: Qifu Li, ; Chuang Wang,
| |
Collapse
|
37
|
In vivo efficacy of verteporfin loaded gold nanorods for combined photothermal/photodynamic colon cancer therapy. Int J Pharm 2022; 625:122134. [PMID: 36007850 DOI: 10.1016/j.ijpharm.2022.122134] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 07/28/2022] [Accepted: 08/18/2022] [Indexed: 02/08/2023]
Abstract
The high incidence of cancer recurrences and the frequent occurrence of multidrug resistance often stem from a poorly selective and inefficient antineoplastic therapy, responsible for the onset of undesired side effects as well. A combination of minimal-invasive approaches could thus be a useful strategy to surmount these shortcomings, achieving a safe and solid cancer therapy. Herein, a multi-therapeutic nanotool was designed by merging the photothermal properties of gold nanorods (AuNRs) with the photodynamic activity of the photosensitizer verteporfin. AuNRs were coated with the natural materials lipoic acid and gellan gum (AuNRs_LA,GG) and subsequently loaded with verteporfin (AuNRs_LA,GG/Vert) producing stable colloidal dispersions. AuNRs_LA,GG/Vert were characterized in terms of stability, size and morphology. The hyperthermia exhibited after NIR excitation (810 nm) was also evaluated to highlight the effect on increasing the drug released profile in intra-tumoral mimicking media, as well as cytotoxicity on human colon cancer cell line (HCT116). In vivo studies on HCT116 murine xenograft models were carried out to prove the ability of AuNRs_LA,GG to arrest the tumor growth via NIR laser-triggered hyperthermia. Furthermore, the complete xenograft depletion was demonstrated upon AuNRs_LA,GG/Vert administration by combined photothermal (PTT) and photodynamic (PDT) effects.
Collapse
|
38
|
Garoffolo G, Casaburo M, Amadeo F, Salvi M, Bernava G, Piacentini L, Chimenti I, Zaccagnini G, Milcovich G, Zuccolo E, Agrifoglio M, Ragazzini S, Baasansuren O, Cozzolino C, Chiesa M, Ferrari S, Carbonaro D, Santoro R, Manzoni M, Casalis L, Raucci A, Molinari F, Menicanti L, Pagano F, Ohashi T, Martelli F, Massai D, Colombo GI, Messina E, Morbiducci U, Pesce M. Reduction of Cardiac Fibrosis by Interference With YAP-Dependent Transactivation. Circ Res 2022; 131:239-257. [PMID: 35770662 DOI: 10.1161/circresaha.121.319373] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Conversion of cardiac stromal cells into myofibroblasts is typically associated with hypoxia conditions, metabolic insults, and/or inflammation, all of which are predisposing factors to cardiac fibrosis and heart failure. We hypothesized that this conversion could be also mediated by response of these cells to mechanical cues through activation of the Hippo transcriptional pathway. The objective of the present study was to assess the role of cellular/nuclear straining forces acting in myofibroblast differentiation of cardiac stromal cells under the control of YAP (yes-associated protein) transcription factor and to validate this finding using a pharmacological agent that interferes with the interactions of the YAP/TAZ (transcriptional coactivator with PDZ-binding motif) complex with their cognate transcription factors TEADs (TEA domain transcription factors), under high-strain and profibrotic stimulation. METHODS We employed high content imaging, 2-dimensional/3-dimensional culture, atomic force microscopy mapping, and molecular methods to prove the role of cell/nuclear straining in YAP-dependent fibrotic programming in a mouse model of ischemia-dependent cardiac fibrosis and in human-derived primitive cardiac stromal cells. We also tested treatment of cells with Verteporfin, a drug known to prevent the association of the YAP/TAZ complex with their cognate transcription factors TEADs. RESULTS Our experiments suggested that pharmacologically targeting the YAP-dependent pathway overrides the profibrotic activation of cardiac stromal cells by mechanical cues in vitro, and that this occurs even in the presence of profibrotic signaling mediated by TGF-β1 (transforming growth factor beta-1). In vivo administration of Verteporfin in mice with permanent cardiac ischemia reduced significantly fibrosis and morphometric remodeling but did not improve cardiac performance. CONCLUSIONS Our study indicates that preventing molecular translation of mechanical cues in cardiac stromal cells reduces the impact of cardiac maladaptive remodeling with a positive effect on fibrosis.
Collapse
Affiliation(s)
- Gloria Garoffolo
- Centro Cardiologico Monzino, IRCCS, Milan, Italy (G.G., M.C., F.A., G.B., L.P., E.Z., S.R., M.C., S.F., R.S., M.M., A.R., G.I.C., M.P.)
| | - Manuel Casaburo
- Centro Cardiologico Monzino, IRCCS, Milan, Italy (G.G., M.C., F.A., G.B., L.P., E.Z., S.R., M.C., S.F., R.S., M.M., A.R., G.I.C., M.P.)
| | - Francesco Amadeo
- Centro Cardiologico Monzino, IRCCS, Milan, Italy (G.G., M.C., F.A., G.B., L.P., E.Z., S.R., M.C., S.F., R.S., M.M., A.R., G.I.C., M.P.)
| | - Massimo Salvi
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy (M.S., D.C., F. Molinari, D.M., U.M.)
| | - Giacomo Bernava
- Centro Cardiologico Monzino, IRCCS, Milan, Italy (G.G., M.C., F.A., G.B., L.P., E.Z., S.R., M.C., S.F., R.S., M.M., A.R., G.I.C., M.P.)
| | - Luca Piacentini
- Centro Cardiologico Monzino, IRCCS, Milan, Italy (G.G., M.C., F.A., G.B., L.P., E.Z., S.R., M.C., S.F., R.S., M.M., A.R., G.I.C., M.P.)
| | - Isotta Chimenti
- Department of Medical Surgical Science and Biotechnology, Sapienza University of Rome (I.C., C.C.).,Mediterranea Cardiocentro, Napoli (I.C.)
| | | | | | - Estella Zuccolo
- Centro Cardiologico Monzino, IRCCS, Milan, Italy (G.G., M.C., F.A., G.B., L.P., E.Z., S.R., M.C., S.F., R.S., M.M., A.R., G.I.C., M.P.)
| | - Marco Agrifoglio
- Dipartimento di Scienze Biomediche, Chirurgiche ed Odontoiatriche, Università di Milano, Milan, Italy (M.A.)
| | - Sara Ragazzini
- Centro Cardiologico Monzino, IRCCS, Milan, Italy (G.G., M.C., F.A., G.B., L.P., E.Z., S.R., M.C., S.F., R.S., M.M., A.R., G.I.C., M.P.)
| | - Otgon Baasansuren
- Faculty of Engineering, Hokkaido University, Sapporo, Japan (O.B., T.O.)
| | - Claudia Cozzolino
- Department of Medical Surgical Science and Biotechnology, Sapienza University of Rome (I.C., C.C.)
| | - Mattia Chiesa
- Centro Cardiologico Monzino, IRCCS, Milan, Italy (G.G., M.C., F.A., G.B., L.P., E.Z., S.R., M.C., S.F., R.S., M.M., A.R., G.I.C., M.P.)
| | - Silvia Ferrari
- Centro Cardiologico Monzino, IRCCS, Milan, Italy (G.G., M.C., F.A., G.B., L.P., E.Z., S.R., M.C., S.F., R.S., M.M., A.R., G.I.C., M.P.)
| | - Dario Carbonaro
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy (M.S., D.C., F. Molinari, D.M., U.M.)
| | - Rosaria Santoro
- Centro Cardiologico Monzino, IRCCS, Milan, Italy (G.G., M.C., F.A., G.B., L.P., E.Z., S.R., M.C., S.F., R.S., M.M., A.R., G.I.C., M.P.)
| | - Martina Manzoni
- Centro Cardiologico Monzino, IRCCS, Milan, Italy (G.G., M.C., F.A., G.B., L.P., E.Z., S.R., M.C., S.F., R.S., M.M., A.R., G.I.C., M.P.)
| | | | - Angela Raucci
- Centro Cardiologico Monzino, IRCCS, Milan, Italy (G.G., M.C., F.A., G.B., L.P., E.Z., S.R., M.C., S.F., R.S., M.M., A.R., G.I.C., M.P.)
| | - Filippo Molinari
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy (M.S., D.C., F. Molinari, D.M., U.M.)
| | | | - Francesca Pagano
- Institute of Biochemistry and Cell Biology, National Council of Research (IBBC-CNR), Monterotondo, Italy (F.P.)
| | - Toshiro Ohashi
- Faculty of Engineering, Hokkaido University, Sapporo, Japan (O.B., T.O.)
| | | | - Diana Massai
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy (M.S., D.C., F. Molinari, D.M., U.M.)
| | - Gualtiero I Colombo
- Centro Cardiologico Monzino, IRCCS, Milan, Italy (G.G., M.C., F.A., G.B., L.P., E.Z., S.R., M.C., S.F., R.S., M.M., A.R., G.I.C., M.P.)
| | - Elisa Messina
- Department of Pediatrics and Infant Neuropsychiatry. Policlinico Umberto I, Sapienza University of Rome (E.M.)
| | - Umberto Morbiducci
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy (M.S., D.C., F. Molinari, D.M., U.M.)
| | - Maurizio Pesce
- Centro Cardiologico Monzino, IRCCS, Milan, Italy (G.G., M.C., F.A., G.B., L.P., E.Z., S.R., M.C., S.F., R.S., M.M., A.R., G.I.C., M.P.)
| |
Collapse
|
39
|
Castro MA, Parson KF, Beg I, Wilkinson MC, Nurmakova K, Levesque I, Voehler MW, Wolfe MS, Ruotolo BT, Sanders CR. Verteporfin is a substrate-selective γ-secretase inhibitor that binds the amyloid precursor protein transmembrane domain. J Biol Chem 2022; 298:101792. [PMID: 35247387 PMCID: PMC8968665 DOI: 10.1016/j.jbc.2022.101792] [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: 12/22/2021] [Revised: 02/22/2022] [Accepted: 02/26/2022] [Indexed: 11/23/2022] Open
Abstract
This work reports substrate-selective inhibition of a protease with broad substrate specificity based on direct binding of a small-molecule inhibitor to the substrate. The target for these studies was γ-secretase protease, which cleaves dozens of different single-span membrane protein substrates, including both the C99 domain of the human amyloid precursor protein and the Notch receptor. Substrate-specific inhibition of C99 cleavage is desirable to reduce production of the amyloid-β polypeptide without inhibiting Notch cleavage, a major source of toxicity associated with broad specificity γ-secretase inhibitors. In order to identify a C99-selective inhibitors of the human γ-secretase, we conducted an NMR-based screen of FDA-approved drugs against C99 in model membranes. From this screen, we identified the small-molecule verteporfin with these properties. We observed that verteporfin formed a direct 1:1 complex with C99, with a KD of 15-47 μM (depending on the membrane mimetic used), and that it did not bind the transmembrane domain of the Notch-1 receptor. Biochemical assays showed that direct binding of verteporfin to C99 inhibits γ-secretase cleavage of C99 with IC50 values in the range of 15-164 μM, while Notch-1 cleavage was inhibited only at higher concentrations, and likely via a mechanism that does not involve binding to Notch-1. This work documents a robust NMR-based approach to discovery of small-molecule binders to single-span membrane proteins and confirmed that it is possible to inhibit γ-secretase in a substrate-specific manner.
Collapse
Affiliation(s)
- Manuel A Castro
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee, USA; Center for Structural Biology, Vanderbilt University, Nashville, Tennessee, USA
| | - Kristine F Parson
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan, USA
| | - Ilyas Beg
- Department of Medicinal Chemistry, University of Kansas, Lawrence, Kansas, USA
| | - Mason C Wilkinson
- Center for Structural Biology, Vanderbilt University, Nashville, Tennessee, USA; Chemical and Physical Biology Program and Center for Structural Biology, Vanderbilt University, Nashville, Tennessee, USA
| | - Kamila Nurmakova
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee, USA
| | - Iliana Levesque
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan, USA
| | - Markus W Voehler
- Center for Structural Biology, Vanderbilt University, Nashville, Tennessee, USA
| | - Michael S Wolfe
- Department of Medicinal Chemistry, University of Kansas, Lawrence, Kansas, USA
| | - Brandon T Ruotolo
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan, USA
| | - Charles R Sanders
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee, USA; Center for Structural Biology, Vanderbilt University, Nashville, Tennessee, USA.
| |
Collapse
|
40
|
Identification of a Quinone Derivative as a YAP/TEAD Activity Modulator from a Repurposing Library. Pharmaceutics 2022; 14:pharmaceutics14020391. [PMID: 35214125 PMCID: PMC8878929 DOI: 10.3390/pharmaceutics14020391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/24/2022] [Accepted: 01/28/2022] [Indexed: 01/25/2023] Open
Abstract
The transcriptional regulators YAP (Yes-associated protein) and TAZ (transcriptional co-activator with PDZ-binding motif) are the major downstream effectors in the Hippo pathway and are involved in cancer progression through modulation of the activity of TEAD (transcriptional enhanced associate domain) transcription factors. To exploit the advantages of drug repurposing in the search of new drugs, we developed a similar approach for the identification of new hits interfering with TEAD target gene expression. In our study, a 27-member in-house library was assembled, characterized, and screened for its cancer cell growth inhibition effect. In a secondary luciferase-based assay, only seven compounds confirmed their specific involvement in TEAD activity. IA5 bearing a p-quinoid structure reduced the cytoplasmic level of phosphorylated YAP and the YAP–TEAD complex transcriptional activity and reduced cancer cell growth. IA5 is a promising hit compound for TEAD activity modulator development.
Collapse
|
41
|
Dos Santos EC, Lalonde-Larue A, Antoniazzi AQ, Barreta MH, Price CA, Dias Gonçalves PB, Portela VM, Zamberlam G. YAP signaling in preovulatory granulosa cells is critical for the functioning of the EGF network during ovulation. Mol Cell Endocrinol 2022; 541:111524. [PMID: 34856345 DOI: 10.1016/j.mce.2021.111524] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 11/24/2021] [Accepted: 11/26/2021] [Indexed: 02/05/2023]
Abstract
Failure to ovulate is a major cause of infertility. The critical pathway that induces ovulation involves the EGF and MAPK phosphorylation, but studies in rodents have suggested that the Hippo activator, YAP, is also involved. It is unknown whether YAP-dependent transcriptional activity is important for the LH- or EGF-induced ovulatory cascade in monovulatory species such as the cow. Using a well-defined preovulatory GC culture system, we employed pharmacological inhibitors to demonstrate that YAP signaling is critical for expression of EGFR and downstream target genes EREG, EGR1 and TNFAIP6. Most importantly, by using an ultrasound guided follicle injection system, we also showed that the classic Hippo signaling inhibitor Verteporfin inhibits GnRH-induced ovulation in vivo in cattle. In conclusion, YAP transcriptional activity is critical for EGF-like cascade induced by LH to promote ovulation in a monovulatory species.
Collapse
Affiliation(s)
- Esdras Corrêa Dos Santos
- Centre de recherche en reproduction et fertilité (CRRF), Faculté de médecine Vétérinaire (FMV), Université de Montréal (UdeM), Canada
| | - Ariane Lalonde-Larue
- Centre de recherche en reproduction et fertilité (CRRF), Faculté de médecine Vétérinaire (FMV), Université de Montréal (UdeM), Canada
| | - Alfredo Quites Antoniazzi
- Laboratory of Biotechnology and Animal Reproduction, BioRep, Veterinary Hospital, Federal University of Santa Maria (UFSM), Santa Maria, RS, 97105-900, Brazil
| | - Marcos Henrique Barreta
- Laboratory of Animal Reproduction Physiology, LAFRA, Federal University of Santa Catarina (UFSC), Curitibanos, SC, 89520-000, Brazil
| | - Christopher A Price
- Centre de recherche en reproduction et fertilité (CRRF), Faculté de médecine Vétérinaire (FMV), Université de Montréal (UdeM), Canada
| | - Paulo Bayard Dias Gonçalves
- Laboratory of Biotechnology and Animal Reproduction, BioRep, Veterinary Hospital, Federal University of Santa Maria (UFSM), Santa Maria, RS, 97105-900, Brazil; Federal University of Pampa (Unipampa), Uruguaiana, RS, 97501-970, Brazil
| | - Valério Marques Portela
- Laboratory of Biotechnology and Animal Reproduction, BioRep, Veterinary Hospital, Federal University of Santa Maria (UFSM), Santa Maria, RS, 97105-900, Brazil
| | - Gustavo Zamberlam
- Centre de recherche en reproduction et fertilité (CRRF), Faculté de médecine Vétérinaire (FMV), Université de Montréal (UdeM), Canada.
| |
Collapse
|
42
|
The YAP/TAZ Signaling Pathway in the Tumor Microenvironment and Carcinogenesis: Current Knowledge and Therapeutic Promises. Int J Mol Sci 2021; 23:ijms23010430. [PMID: 35008857 PMCID: PMC8745604 DOI: 10.3390/ijms23010430] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/23/2021] [Accepted: 12/24/2021] [Indexed: 12/14/2022] Open
Abstract
The yes-associated protein (YAP) and the transcriptional coactivator with PDZ-binding motif (TAZ) are transcriptional coactivators, members of the Hippo signaling pathway, which play a critical role in cell growth regulation, embryonic development, regeneration, proliferation, and cancer origin and progression. The mechanism involves the nuclear binding of the un-phosphorylated YAP/TAZ complex to release the transcriptional enhanced associate domain (TEAD) from its repressors. The active ternary complex is responsible for the aforementioned biological effects. Overexpression of YAP/TAZ has been reported in cancer stem cells and tumor resistance. The resistance involves chemotherapy, targeted therapy, and immunotherapy. This review provides an overview of YAP/TAZ pathways’ role in carcinogenesis and tumor microenvironment. Potential therapeutic alternatives are also discussed.
Collapse
|
43
|
Barrette AM, Ronk H, Joshi T, Mussa Z, Mehrotra M, Bouras A, Nudelman G, Jesu Raj JG, Bozec D, Lam W, Houldsworth J, Yong R, Zaslavsky E, Hadjipanayis CG, Birtwistle MR, Tsankova NM. Anti-invasive efficacy and survival benefit of the YAP-TEAD inhibitor Verteporfin in preclinical glioblastoma models. Neuro Oncol 2021; 24:694-707. [PMID: 34657158 DOI: 10.1093/neuonc/noab244] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Glioblastoma (GBM) remains a largely incurable disease as current therapy fails to target the invasive nature of GBM growth in disease progression and recurrence. Here we use the FDA-approved drug and small molecule Hippo inhibitor Verteporfin to target YAP-TEAD activity, known to mediate convergent aspects of tumor invasion/metastasis, and assess the drug's efficacy and survival benefit in GBM models. METHODS Up to eight low-passage patient-derived GBM cell lines with distinct genomic drivers, including three primary/recurrent pairs, were treated with Verteporfin or vehicle to assess in-vitro effects on proliferation, migration, YAP-TEAD activity, and transcriptomics. Patient-derived orthotopic xenograft models (PDX) were used to assess Verteporfin's brain penetrance and effects on tumor burden and survival. RESULTS Verteporfin treatment disturbed YAP/TAZ-TEAD activity; disrupted transcriptome signatures related to invasion, epithelial-to-mesenchymal, and proneural-to-mesenchymal transition, phenocopying TEAD1-knockout effects; and impaired tumor migration/invasion dynamics across primary and recurrent GBM lines. In an aggressive orthotopic PDX GBM model, short-term Verteporfin treatment consistently diminished core and infiltrative tumor burden, which was associated with decreased tumor expression of Ki67, nuclear YAP, TEAD1, and TEAD-associated targets EGFR, CDH2 and ITGB1. Finally, long-term Verteporfin treatment appeared non-toxic and conferred survival benefit compared to vehicle in two PDX models: as monotherapy in primary (de-novo) GBM and in combination with Temozolomide chemoradiation in recurrent GBM, where VP treatment associated with increased MGMT methylation. CONCLUSIONS We demonstrate combined anti-invasive and anti-proliferative efficacy for Verteporfin with survival benefit in preclinical GBM models, indicating potential therapeutic value of this already FDA-approved drug if repurposed for glioblastoma patients.
Collapse
Affiliation(s)
- Anne Marie Barrette
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Neurosurgery, Stanford University, Stanford, CA, USA
| | - Halle Ronk
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Tanvi Joshi
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Zarmeen Mussa
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Meenakshi Mehrotra
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alexandros Bouras
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - German Nudelman
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Joe G Jesu Raj
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Dominique Bozec
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - William Lam
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jane Houldsworth
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Raymund Yong
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Elena Zaslavsky
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Marc R Birtwistle
- Department of Chemical and Biomolecular Engineering, Clemson University, Clemson, South Carolina, USA
| | - Nadejda M Tsankova
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
44
|
Wei X, Lou H, Zhou D, Jia Y, Li H, Huang Q, Ma J, Yang Z, Sun C, Meng Y, Xu S, Yang X, Li X, Ji T, Guo Z, Gao Q. TAGLN mediated stiffness-regulated ovarian cancer progression via RhoA/ROCK pathway. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:292. [PMID: 34538264 PMCID: PMC8451140 DOI: 10.1186/s13046-021-02091-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 08/31/2021] [Indexed: 12/14/2022]
Abstract
Background Ovarian cancer (OC) progression is an unmet medical challenge. Since omental metastases were palpated harder than their primary counterparts during cytoreductive surgery of patients with epithelial ovarian cancer (EOC), we were inspired to investigate OC progression from the perspective of biomechanics. Methods Atomic Force Microscope (AFM) was used to measure the Young’s modulus of tissues. The collagen-coated polyacrylamide hydrogel (PA gel) system was prepared to mimic the soft and stiff substrates in vitro. The effect of TAGLN was evaluated both in vitro and in vivo using transwell assay, immunofluorescence, western blot analysis and immunohistochemistry. Results We quantitatively confirmed that omental metastases were stiffer and more abundant in desmoplasia compared with paired primary tumors, and further demonstrated that matrix stiffness could notably regulate OC progression. Remarkably, TAGLN, encoding an actin cross-linking/gelling protein, was identified as a potent mechanosensitive gene that could form a regulation loop with Src activation reacting to environmental stiffness, thus mediating stiffness-regulated OC progression through regulating RhoA/ROCK pathway. Conclusions These data demonstrate that targeting extra-cellular matrix (ECM) stiffness could probably hamper OC progression, and of note, targeting TAGLN might provide promising clinical therapeutic value for OC therapy. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-021-02091-6.
Collapse
Affiliation(s)
- Xiao Wei
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Anv, Wuhan, People's Republic of China
| | - Hua Lou
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Anv, Wuhan, People's Republic of China.,Department of Gynecology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Dongchen Zhou
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Anv, Wuhan, People's Republic of China
| | - Yijuan Jia
- Department of Obstetrics and Gynecology, Wuhan First Hospital, Wuhan, People's Republic of China
| | - Huayi Li
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Anv, Wuhan, People's Republic of China
| | - Quanfu Huang
- Department of Thoracic Surgery, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Jingjing Ma
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Anv, Wuhan, People's Republic of China
| | - Zongyuan Yang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Anv, Wuhan, People's Republic of China
| | - Chaoyang Sun
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Anv, Wuhan, People's Republic of China
| | - Yunchong Meng
- Department of Thoracic Surgery, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Sen Xu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Anv, Wuhan, People's Republic of China
| | - Xin Yang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Anv, Wuhan, People's Republic of China
| | - Xiaoting Li
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Anv, Wuhan, People's Republic of China
| | - Teng Ji
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Anv, Wuhan, People's Republic of China
| | - Zhongzhen Guo
- Shenzhen Dapeng New District Maternity & Child Health Hospital Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Qinglei Gao
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Anv, Wuhan, People's Republic of China.
| |
Collapse
|
45
|
Fan Y, Sun Q, Li X, Feng J, Ao Z, Li X, Wang J. Substrate Stiffness Modulates the Growth, Phenotype, and Chemoresistance of Ovarian Cancer Cells. Front Cell Dev Biol 2021; 9:718834. [PMID: 34504843 PMCID: PMC8421636 DOI: 10.3389/fcell.2021.718834] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 07/27/2021] [Indexed: 01/01/2023] Open
Abstract
Mechanical factors in the tumor microenvironment play an important role in response to a variety of cellular activities in cancer cells. Here, we utilized polyacrylamide hydrogels with varying physical parameters simulating tumor and metastatic target tissues to investigate the effect of substrate stiffness on the growth, phenotype, and chemotherapeutic response of ovarian cancer cells (OCCs). We found that increasing the substrate stiffness promoted the proliferation of SKOV-3 cells, an OCC cell line. This proliferation coincided with the nuclear translocation of the oncogene Yes-associated protein. Additionally, we found that substrate softening promoted elements of epithelial-mesenchymal transition (EMT), including mesenchymal cell shape changes, increase in vimentin expression, and decrease in E-cadherin and β-catenin expression. Growing evidence demonstrates that apart from contributing to cancer initiation and progression, EMT can promote chemotherapy resistance in ovarian cancer cells. Furthermore, we evaluated tumor response to standard chemotherapeutic drugs (cisplatin and paclitaxel) and found antiproliferation effects to be directly proportional to the stiffness of the substrate. Nanomechanical studies based on atomic force microscopy (AFM) have revealed that chemosensitivity and chemoresistance are related to cellular mechanical properties. The results of cellular elastic modulus measurements determined by AFM demonstrated that Young's modulus of SKOV-3 cells grown on soft substrates was less than that of cells grown on stiff substrates. Gene expression analysis of SKOV-3 cells showed that mRNA expression can be greatly affected by substrate stiffness. Finally, immunocytochemistry analyses revealed an increase in multidrug resistance proteins, namely, ATP binding cassette subfamily B member 1 and member 4 (ABCB1 and ABCB4), in the cells grown on the soft gel resulting in resistance to chemotherapeutic drugs. In conclusion, our study may help in identification of effective targets for cancer therapy and improve our understanding of the mechanisms of cancer progression and chemoresistance.
Collapse
Affiliation(s)
- Yali Fan
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Quanmei Sun
- Chinese Academy of Sciences (CAS) Center for Excellence in Nanoscience, National Centre for Nanoscience and Technology, Beijing, China
| | - Xia Li
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China.,Hospital of Beijing Forestry University, Beijing Forestry University, Beijing, China
| | - Jiantao Feng
- Artemisinin Research Center, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhuo Ao
- Chinese Academy of Sciences (CAS) Center for Excellence in Nanoscience, National Centre for Nanoscience and Technology, Beijing, China
| | - Xiang Li
- Chinese Academy of Sciences (CAS) Center for Excellence in Nanoscience, National Centre for Nanoscience and Technology, Beijing, China
| | - Jiandong Wang
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
46
|
Hasegawa T, Sugihara T, Hoshino Y, Tarumoto R, Matsuki Y, Kanda T, Takata T, Nagahara T, Matono T, Isomoto H. Photosensitizer verteporfin inhibits the growth of YAP- and TAZ-dominant gastric cancer cells by suppressing the anti-apoptotic protein Survivin in a light-independent manner. Oncol Lett 2021; 22:703. [PMID: 34457058 PMCID: PMC8358589 DOI: 10.3892/ol.2021.12964] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 07/07/2021] [Indexed: 12/11/2022] Open
Abstract
Yes-associated protein (YAP) positivity indicates a poor prognosis in gastric cancer. Transcriptional co-activator with a PDZ-binding domain (TAZ), a YAP paralog, is highly expressed in gastric signet ring cell carcinoma. Verteporfin (VP), a clinical photosensitizer, was recently shown to inhibit YAP/TAZ. In the present study, the therapeutic potential of VP treatment was explored using two gastric cancer cell lines: MKN-45 (TAZ-dominant) and MKN-74 (YAP-dominant). Cell proliferation was evaluated by MTS assay. Vascular mimicry was evaluated by the tube formation assay. Gene and protein expression levels of YAP/TAZ downstream effectors [such as Survivin, Cysteine-rich angiogenic inducer 61 (CYR61), and connective tissue growth factor (CTGF)] were measured. YAP or TAZ localization was evaluated by immunofluorescence. Cell death was assessed by immunofluorescent staining of Annexin V. YAP and TAZ expression were knocked down by small interfering RNA. The current results demonstrate that MKN-45, a poorly differentiated TAZ-dominant gastric cancer cell line, was more sensitive to VP than MKN-74, a moderately differentiated YAP-dominant gastric cancer cell line. VP changed the localization of YAP/TAZ, promoted its degradation and significantly decreased the protein level of Survivin in both cell lines. Cell death was induced by VP treatment in a dose-dependent manner. Vascular mimicry was inhibited in both cell lines. Proliferation in both cell lines decreased in response to YAP/TAZ knockdown. The present study indicated that VP has potential as a therapeutic agent in YAP- and TAZ-dominant gastric cancers due to its ability to suppress the anti-apoptotic protein Survivin via inhibition of YAP and TAZ.
Collapse
Affiliation(s)
- Takashi Hasegawa
- Division of Medicine and Clinical Science, Department of Gastroenterology and Nephrology, School of Medicine, Faculty of Medicine, Tottori University, Yonago 683-8504, Japan
| | - Takaaki Sugihara
- Division of Medicine and Clinical Science, Department of Gastroenterology and Nephrology, School of Medicine, Faculty of Medicine, Tottori University, Yonago 683-8504, Japan
| | - Yoshiki Hoshino
- Division of Medicine and Clinical Science, Department of Gastroenterology and Nephrology, School of Medicine, Faculty of Medicine, Tottori University, Yonago 683-8504, Japan
| | - Ryohei Tarumoto
- Division of Medicine and Clinical Science, Department of Gastroenterology and Nephrology, School of Medicine, Faculty of Medicine, Tottori University, Yonago 683-8504, Japan
| | - Yukako Matsuki
- Division of Medicine and Clinical Science, Department of Gastroenterology and Nephrology, School of Medicine, Faculty of Medicine, Tottori University, Yonago 683-8504, Japan
| | - Tsutomu Kanda
- Division of Medicine and Clinical Science, Department of Gastroenterology and Nephrology, School of Medicine, Faculty of Medicine, Tottori University, Yonago 683-8504, Japan
| | - Tomoaki Takata
- Division of Medicine and Clinical Science, Department of Gastroenterology and Nephrology, School of Medicine, Faculty of Medicine, Tottori University, Yonago 683-8504, Japan
| | - Takakazu Nagahara
- Division of Medicine and Clinical Science, Department of Gastroenterology and Nephrology, School of Medicine, Faculty of Medicine, Tottori University, Yonago 683-8504, Japan
| | - Tomomitsu Matono
- Division of Medicine and Clinical Science, Department of Gastroenterology and Nephrology, School of Medicine, Faculty of Medicine, Tottori University, Yonago 683-8504, Japan
| | - Hajime Isomoto
- Division of Medicine and Clinical Science, Department of Gastroenterology and Nephrology, School of Medicine, Faculty of Medicine, Tottori University, Yonago 683-8504, Japan
| |
Collapse
|
47
|
Verduci L, Tarcitano E, Strano S, Yarden Y, Blandino G. CircRNAs: role in human diseases and potential use as biomarkers. Cell Death Dis 2021; 12:468. [PMID: 33976116 PMCID: PMC8113373 DOI: 10.1038/s41419-021-03743-3] [Citation(s) in RCA: 191] [Impact Index Per Article: 63.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 04/18/2021] [Accepted: 04/19/2021] [Indexed: 02/07/2023]
Abstract
Circular RNAs (circRNAs) are a class of endogenous RNAs characterized by a covalent loop structure. In comparison to other types of RNAs, the abundance of circRNAs is relatively low but due to the circular configuration, their stability is very high. In addition, circRNAs display high degree of tissue specificity. The sponging activity of circRNAs toward microRNAs is the best-described mode of action of circRNAs. However, the ability of circRNAs to bind with specific proteins, as well as to encode short proteins, propose alternative functions. This review introduces the biogenesis of circRNAs and summarizes the roles played by circRNAs in human diseases. These include examples of their functional roles in several organ-specific cancers, such as head and neck and breast and lung cancers. In addition, we review potential functions of circRNAs in diabetes, cardiovascular, and neurodegenerative diseases. Recently, a growing number of studies have demonstrated involvement of circRNAs in a wide spectrum of signaling molecular pathways, but at the same time many different and controversial views on circRNAs role and function are emerging. We conclude by offering cellular homeostasis generated by networks comprising circular RNAs, other non-coding RNAs and RNA-binding proteins. Accordingly, it is predictable that circRNAs, due to their highly stable nature and remarkable tissue specificity, will emerge as reliable biomarkers of disease course and treatment efficacy.
Collapse
Affiliation(s)
- Lorena Verduci
- Unit of Oncogenomic and Epigenetic, Department of Research, Advanced Diagnostic, and Technological Innovation, IRCCS, Regina Elena National Cancer Institute, Rome, Italy
| | - Emilio Tarcitano
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Sabrina Strano
- SAFU Unit, Department of Research, Advanced Diagnostic, and Technological Innovation, IRCCS, Regina Elena National Cancer Institute, Rome, Italy
| | - Yosef Yarden
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel.
| | - Giovanni Blandino
- Unit of Oncogenomic and Epigenetic, Department of Research, Advanced Diagnostic, and Technological Innovation, IRCCS, Regina Elena National Cancer Institute, Rome, Italy.
| |
Collapse
|
48
|
Walther BK, Rajeeva Pandian NK, Gold KA, Kiliç ES, Sama V, Gu J, Gaharwar AK, Guiseppi-Elie A, Cooke JP, Jain A. Mechanotransduction-on-chip: vessel-chip model of endothelial YAP mechanobiology reveals matrix stiffness impedes shear response. LAB ON A CHIP 2021; 21:1738-1751. [PMID: 33949409 PMCID: PMC9761985 DOI: 10.1039/d0lc01283a] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Endothelial mechanobiology is a key consideration in the progression of vascular dysfunction, including atherosclerosis. However mechanistic connections between the clinically associated physical stimuli, vessel stiffness and shear stress, and how they interact to modulate plaque progression remain incompletely characterized. Vessel-chip systems are excellent candidates for modeling vascular mechanobiology as they may be engineered from the ground up, guided by the mechanical parameters present in human arteries and veins, to recapitulate key features of the vasculature. Here, we report extensive validation of a vessel-chip model of endothelial yes-associated protein (YAP) mechanobiology, a protein sensitive to both matrix stiffness and shearing forces and, importantly, implicated in atherosclerotic progression. Our model captures the established endothelial mechanoresponse, with endothelial alignment, elongation, reduction of adhesion molecules, and YAP cytoplasmic retention under high laminar shear. Conversely, we observed disturbed morphology, inflammation, and nuclear partitioning under low, high, and high oscillatory shear. Examining targets of YAP transcriptional co-activation, connective tissue growth factor (CTGF) is strongly downregulated by high laminar shear, whereas it is strongly upregulated by low shear or oscillatory flow. Ankyrin repeat domain 1 (ANKRD1) is only upregulated by high oscillatory shear. Verteporfin inhibition of YAP reduced the expression of CTGF but did not affect ANKRD1. Lastly, substrate stiffness modulated the endothelial shear mechanoresponse. Under high shear, softer substrates showed the lowest nuclear localization of YAP whereas stiffer substrates increased nuclear localization. Low shear strongly increased nuclear localization of YAP across stiffnesses. Together, we have validated a model of endothelial mechanobiology and describe a clinically relevant biological connection between matrix stiffness, shear stress, and endothelial activation via YAP mechanobiology.
Collapse
Affiliation(s)
- Brandon K Walther
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas 77843, USA. and Department of Cardiovascular Sciences, Houston Methodist Institute for Academic Medicine, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, Texas 77030, USA.
| | | | - Karli A Gold
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas 77843, USA.
| | - Ecem S Kiliç
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas 77843, USA.
| | - Vineeth Sama
- Department of Biomedical Engineering, Clemson University, Clemson, South Carolina 29634, USA.
| | - Jianhua Gu
- Department of Cardiovascular Sciences, Houston Methodist Institute for Academic Medicine, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, Texas 77030, USA.
| | - Akhilesh K Gaharwar
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas 77843, USA. and Department of Materials Science, Texas A&M University, College Station, Texas 77843, USA
| | - Anthony Guiseppi-Elie
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas 77843, USA. and Department of Cardiovascular Sciences, Houston Methodist Institute for Academic Medicine, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, Texas 77030, USA. and ABTECH Scientific, Inc., Biotechnology Research Park, 800 East Leigh Street, Richmond, Virginia 23219, USA and Department of Biomedical Engineering, Anderson University, Anderson, South Carolina 29621, USA.
| | - John P Cooke
- Department of Cardiovascular Sciences, Houston Methodist Institute for Academic Medicine, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, Texas 77030, USA.
| | - Abhishek Jain
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas 77843, USA. and Department of Cardiovascular Sciences, Houston Methodist Institute for Academic Medicine, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, Texas 77030, USA. and Department of Medical Physiology, College of Medicine, Texas A&M Health Science Center, Bryan, TX 77807, USA
| |
Collapse
|
49
|
Tian Y, Zhang MY, Zhao AH, Kong L, Wang JJ, Shen W, Li L. Single-cell transcriptomic profiling provides insights into the toxic effects of Zearalenone exposure on primordial follicle assembly. Am J Cancer Res 2021; 11:5197-5213. [PMID: 33859742 PMCID: PMC8039963 DOI: 10.7150/thno.58433] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 02/21/2021] [Indexed: 12/15/2022] Open
Abstract
Rationale: Zearalenone (ZEN), a pollutant in our daily diet, seriously threatens the reproductive health of humans and animals. The primordial follicle (PF) assembly in the mouse occurs during the perinatal period, which determines the whole ovarian reserve in reproductive lifespan. In the current investigation, we administered ZEN orally to perinatal mice from 16.5 days post coitum (dpc) to postnatal day 3 (PD3), and single-cell RNA sequencing (scRNA-seq) was performed on PD0 and PD3 ovarian tissues in the offspring to check ZEN toxic to primordial follicle formation at the single cell level. Methods: Ovarian tissues (in vivo) were examined by single cell RNA sequencing analysis, Immunostaining, and Western blotting. Ovarian tissues (in vitro) were examined by qRT-PCR, Immunostaining, and Western blotting. Results: We found that ZEN exposure altered the developmental trajectory of both germ cells and granulosa cells. Furthermore, after establishing the cell-cell communication network between germ cells and granulosa cells, we found that this was perturbed by ZEN exposure, especially during the Hippo signaling pathway. Conclusions: This study showed that ZEN affected the status of germ cells and granulosa cells through the Hippo signaling pathway and blocked the assembly of PFs. This research contributes to our deeper understanding of the mechanisms of toxicity in different cell types and the disruption of normal intercellular signaling by ZEN exposure.
Collapse
|
50
|
Zhuang Q, Li F, Liu J, Wang H, Tian Y, Zhang Z, Wang F, Zhao Z, Chen J, Wu H. Nuclear exclusion of YAP exacerbates podocyte apoptosis and disease progression in Adriamycin-induced focal segmental glomerulosclerosis. J Transl Med 2021; 101:258-270. [PMID: 33203894 PMCID: PMC7815513 DOI: 10.1038/s41374-020-00503-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Revised: 10/09/2020] [Accepted: 10/16/2020] [Indexed: 01/19/2023] Open
Abstract
Focal segmental glomerulosclerosis (FSGS) is a chronic glomerular disease with poor clinical outcomes. Podocyte loss via apoptosis is one important mechanism underlying the pathogenesis of FSGS. Recently, Yes-associated-protein (YAP), a key downstream protein in the Hippo pathway, was identified as an activator for multiple gene transcriptional factors in the nucleus to control cell proliferation and apoptosis. To investigate the potential role of YAP in the progression of FSGS, we examined kidney samples from patients with minimal change disease or FSGS and found that increases in podocyte apoptosis is positively correlated with the cytoplasmic distribution of YAP in human FSGS. Utilizing an established mT/mG transgenic mouse model and primary cultured podocytes, we found that YAP was distributed uniformly in nucleus and cytoplasm in the podocytes of control animals. Adriamycin treatment induced gradual nuclear exclusion of YAP with enhanced phospho-YAP/YAP ratio, accompanied by the induction of podocyte apoptosis both in vivo and in vitro. Moreover, we used verteporfin to treat an Adriamycin-induced FSGS mouse model, and found YAP inhibition by verteporfin induced nuclear exclusion of YAP, thus increasing podocyte apoptosis and accelerating disease progression. Therefore, our findings suggest that YAP nuclear distribution and activation in podocytes is an important endogenous anti-apoptotic mechanism during the progression of FSGS.
Collapse
Affiliation(s)
- Qiyuan Zhuang
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, China
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Fang Li
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, China
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jun Liu
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongyu Wang
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yuchen Tian
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Zhigang Zhang
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Feng Wang
- Department of Nephrology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Zhonghua Zhao
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Jianchun Chen
- Division of Nephrology in Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA.
| | - Huijuan Wu
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, China.
| |
Collapse
|