1
|
Liu SF, Kucherenko MM, Sang P, Li Q, Yao J, Nambiar Veetil N, Gransar T, Alesutan I, Voelkl J, Salinas G, Grune J, Simmons S, Knosalla C, Kuebler WM. RUNX2 is stabilised by TAZ and drives pulmonary artery calcification and lung vascular remodelling in pulmonary hypertension due to left heart disease. Eur Respir J 2024; 64:2300844. [PMID: 39542509 DOI: 10.1183/13993003.00844-2023] [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: 05/19/2023] [Accepted: 07/13/2024] [Indexed: 11/17/2024]
Abstract
BACKGROUND Calcification is common in chronic vascular disease, yet its role in pulmonary hypertension due to left heart disease is unknown. Here, we probed for the role of runt-related transcription factor-2 (RUNX2), a master transcription factor in osteogenesis, and its regulation by the HIPPO pathway transcriptional coactivator with PDZ-binding motif (TAZ) in the osteogenic reprogramming of pulmonary artery smooth muscle cells and vascular calcification in patients with pulmonary hypertension due to left heart disease. We similarly examined its role using an established rat model of pulmonary hypertension due to left heart disease induced by supracoronary aortic banding. METHODS Pulmonary artery samples were collected from patients and rats with pulmonary hypertension due to left heart disease. Genome-wide RNA sequencing was performed, and pulmonary artery calcification assessed. Osteogenic signalling via TAZ and RUNX2 was delineated by protein biochemistry. In vivo, the therapeutic potential of RUNX2 or TAZ inhibition by CADD522 or verteporfin was tested in the rat model. RESULTS Gene ontology term analysis identified significant enrichment in ossification and osteoblast differentiation genes, including RUNX2, in pulmonary arteries of patients and lungs of rats with pulmonary hypertension due to left heart disease. Pulmonary artery calcification was evident in both patients and rats. Both TAZ and RUNX2 were upregulated and activated in pulmonary artery smooth muscle cells of patients and rats. Co-immunoprecipitation revealed a direct interaction of RUNX2 with TAZ in pulmonary artery smooth muscle cells. TAZ inhibition or knockdown decreased RUNX2 abundance due to accelerated RUNX2 protein degradation rather than reduced de novo synthesis. Inhibition of either TAZ or RUNX2 attenuated pulmonary artery calcification, distal lung vascular remodelling and pulmonary hypertension development in the rat model. CONCLUSION Pulmonary hypertension due to left heart disease is associated with pulmonary artery calcification that is driven by TAZ-dependent stabilisation of RUNX2, causing osteogenic reprogramming of pulmonary artery smooth muscle cells. The TAZ-RUNX2 axis may present a therapeutic target in pulmonary hypertension due to left heart disease.
Collapse
Affiliation(s)
- Shao-Fei Liu
- Institute for Physiology, Charité - Universitätsmedizin Berlin, corporate member of the Free University Berlin and the Humboldt University Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- These authors contributed equally to the study
| | - Mariya M Kucherenko
- Institute for Physiology, Charité - Universitätsmedizin Berlin, corporate member of the Free University Berlin and the Humboldt University Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of the Free University Berlin and the Humboldt University Berlin, Berlin, Germany
- These authors contributed equally to the study
| | - Pengchao Sang
- Institute for Physiology, Charité - Universitätsmedizin Berlin, corporate member of the Free University Berlin and the Humboldt University Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
| | - Qiuhua Li
- Institute for Physiology, Charité - Universitätsmedizin Berlin, corporate member of the Free University Berlin and the Humboldt University Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Juquan Yao
- Institute for Physiology, Charité - Universitätsmedizin Berlin, corporate member of the Free University Berlin and the Humboldt University Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Netra Nambiar Veetil
- Institute for Physiology, Charité - Universitätsmedizin Berlin, corporate member of the Free University Berlin and the Humboldt University Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of the Free University Berlin and the Humboldt University Berlin, Berlin, Germany
| | - Tara Gransar
- Institute for Physiology, Charité - Universitätsmedizin Berlin, corporate member of the Free University Berlin and the Humboldt University Berlin, Berlin, Germany
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
| | - Ioana Alesutan
- Institute of Physiology and Pathophysiology, Johannes Kepler University Linz, Linz, Austria
| | - Jakob Voelkl
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- Institute of Physiology and Pathophysiology, Johannes Kepler University Linz, Linz, Austria
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Gabriela Salinas
- NGS - Integrative Genomics Core Unit (NIG), Institute of Pathology, University Medical Center Göttingen (UMG), Göttingen, Germany
| | - Jana Grune
- Institute for Physiology, Charité - Universitätsmedizin Berlin, corporate member of the Free University Berlin and the Humboldt University Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Szandor Simmons
- Institute for Physiology, Charité - Universitätsmedizin Berlin, corporate member of the Free University Berlin and the Humboldt University Berlin, Berlin, Germany
| | - Christoph Knosalla
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of the Free University Berlin and the Humboldt University Berlin, Berlin, Germany
- These authors share the last authorship
| | - Wolfgang M Kuebler
- Institute for Physiology, Charité - Universitätsmedizin Berlin, corporate member of the Free University Berlin and the Humboldt University Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- DZL (German Centre for Lung Research), partner site Berlin, Berlin, Germany
- The Keenan Research Centre for Biomedical Science at St. Michael's, Toronto, ON, Canada
- Departements of Surgery and Physiology, University of Toronto, Toronto, ON, Canada
- These authors share the last authorship
| |
Collapse
|
2
|
Mohammadpour S, Torshizi Esfahani A, Sarpash S, Vakili F, Zafarjafarzadeh N, Mashaollahi A, Pardakhtchi A, Nazemalhosseini-Mojarad E. Hippo Signaling Pathway in Colorectal Cancer: Modulation by Various Signals and Therapeutic Potential. Anal Cell Pathol (Amst) 2024; 2024:5767535. [PMID: 39431199 PMCID: PMC11489006 DOI: 10.1155/2024/5767535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 07/07/2024] [Accepted: 08/19/2024] [Indexed: 10/22/2024] Open
Abstract
Colorectal cancer (CRC) stands as a significant global health issue, marked by elevated occurrence and mortality statistics. Despite the availability of various treatments, including chemotherapy, radiotherapy, and targeted therapy, CRC cells often exhibit resistance to these interventions. As a result, it is imperative to identify the disease at an earlier stage and enhance the response to treatment by acquiring a deeper comprehension of the processes driving tumor formation, aggressiveness, metastasis, and resistance to therapy. The Hippo pathway plays a critical role in facilitating the initiation of tumorigenesis and frequently experiences disruption within CRC because of genetic mutations and modified expression in its fundamental constituents. Targeting upstream regulators or core Hippo pathway components may provide innovative therapeutic strategies for modulating Hippo signaling dysfunction in CRC. To advance novel therapeutic techniques for CRC, it is imperative to grasp the involvement of the Hippo pathway in CRC and its interaction with alternate signaling pathways, noncoding RNAs, gut microbiota, and the immune microenvironment. This review seeks to illuminate the function and control of the Hippo pathway in CRC, ultimately aiming to unearth innovative therapeutic methodologies for addressing this ailment.
Collapse
Affiliation(s)
- Somayeh Mohammadpour
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir Torshizi Esfahani
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - SeyedKasra Sarpash
- Department of Cellular and Molecular Biology, Faculty of Advanced Science and Technology, Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Fatemeh Vakili
- Department of Cellular and Molecular Biology, Faculty of Advanced Science and Technology, Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Nikta Zafarjafarzadeh
- Department of Cellular and Molecular Biology, Faculty of Advanced Science and Technology, Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Amirhesam Mashaollahi
- Department of Cellular and Molecular Biology, Faculty of Advanced Science and Technology, Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Ali Pardakhtchi
- Department of Cellular and Molecular Biology, Faculty of Advanced Science and Technology, Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Ehsan Nazemalhosseini-Mojarad
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
3
|
Wang X, Wang S, Mu H, Yang C, Dong W, Wang X, Wang J. Macrophage-derived amphiregulin promoted the osteogenic differentiation of chondrocytes through EGFR/Yap axis and TGF-β activation. Bone 2024; 190:117275. [PMID: 39383984 DOI: 10.1016/j.bone.2024.117275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 07/15/2024] [Accepted: 10/04/2024] [Indexed: 10/11/2024]
Abstract
Endochondral ossification represents a crucial biological process in skeletal development and bone defect repair. Macrophages, recognized as key players in the immune system, are now acknowledged for their substantial role in promoting endochondral ossification within cartilage. Concurrently, the epidermal growth factor receptor (EGFR) ligand amphiregulin (Areg) has been documented for its contributory role in restoring bone tissue homeostasis post-injury. However, the mechanism by which macrophage-secreted Areg facilitates bone repair remains elusive. In this study, the induction of macrophage depletion through in vivo administration of clodronate liposomes was employed in a standard open tibial fracture mouse model to assess bone healing using micro-computed tomography (micro-CT) analysis, histomorphology, and ELISA serum evaluations. The investigation revealed sustained expression of Areg during the fracture healing period in wild-type mice. Macrophage depletion significantly reduced the number of macrophages on the local bone surface and vital organs. This reduction led to diminished Areg secretion, decreased collagen production, and delayed fracture healing. However, histological and micro-CT assessments at 7 and 21 days post-local Areg treatment exhibited a marked improvement of bone healing compared to the vehicle control. In vitro studies demonstrated an increase of Areg secretion by the Raw264.7 cells upon ATP stimulation. Indirect co-culture of Raw264.7 and ATDC5 cells indicated that Areg overexpression enhanced the osteogenic potential of chondrocytes, and vice versa. This osteogenic promotion was attributed to Areg's activation of the membrane receptor EGFR in the ATDC5 cell line, the enhanced phosphorylation of transcription factor Yap, and the facilitation of the expression of bioactive TGF-β by chondrocytes. Collectively, this research elucidates the direct mechanistic effects of macrophage-secreted Areg in promoting bone homeostasis following bone injury.
Collapse
Affiliation(s)
- Xinyi Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China
| | - Shuo Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China
| | - Hailin Mu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China
| | - Chang Yang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China
| | - Wei Dong
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China
| | - Xinru Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China
| | - Jaiwei Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China.
| |
Collapse
|
4
|
Venugopal S, Dan Q, Sri Theivakadadcham VS, Wu B, Kofler M, Layne MD, Connelly KA, Rzepka MF, Friedberg MK, Kapus A, Szászi K. Regulation of the RhoA exchange factor GEF-H1 by profibrotic stimuli through a positive feedback loop involving RhoA, MRTF, and Sp1. Am J Physiol Cell Physiol 2024; 327:C387-C402. [PMID: 38912734 DOI: 10.1152/ajpcell.00088.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 06/03/2024] [Accepted: 06/14/2024] [Indexed: 06/25/2024]
Abstract
RhoA and its effectors, the transcriptional coactivators myocardin-related transcription factor (MRTF) and serum response factor (SRF), control epithelial phenotype and are indispensable for profibrotic epithelial reprogramming during fibrogenesis. Context-dependent control of RhoA and fibrosis-associated changes in its regulators, however, remain incompletely characterized. We previously identified the guanine nucleotide exchange factor GEF-H1 as a central mediator of RhoA activation in renal tubular cells exposed to inflammatory or fibrotic stimuli. Here we found that GEF-H1 expression and phosphorylation were strongly elevated in two animal models of fibrosis. In the Unilateral Ureteral Obstruction mouse kidney fibrosis model, GEF-H1 was upregulated predominantly in the tubular compartment. GEF-H1 was also elevated and phosphorylated in a rat pulmonary artery banding (PAB) model of right ventricular fibrosis. Prolonged stimulation of LLC-PK1 tubular cells with tumor necrosis factor (TNF)-α or transforming growth factor (TGF)-β1 increased GEF-H1 expression and activated a luciferase-coupled GEF-H1 promoter. Knockdown and overexpression studies revealed that these effects were mediated by RhoA, cytoskeleton remodeling, and MRTF, indicative of a positive feedback cycle. Indeed, silencing endogenous GEF-H1 attenuated activation of the GEF-H1 promoter. Of importance, inhibition of MRTF using CCG-1423 prevented GEF-H1 upregulation in both animal models. MRTF-dependent increase in GEF-H1 was prevented by inhibition of the transcription factor Sp1, and mutating putative Sp1 binding sites in the GEF-H1 promoter eliminated its MRTF-dependent activation. As the GEF-H1/RhoA axis is key for fibrogenesis, this novel MRTF/Sp1-dependent regulation of GEF-H1 abundance represents a potential target for reducing renal and cardiac fibrosis.NEW & NOTEWORTHY We show that expression of the RhoA regulator GEF-H1 is upregulated in tubular cells exposed to fibrogenic cytokines and in animal models of kidney and heart fibrosis. We identify a pathway wherein GEF-H1/RhoA-dependent MRTF activation through its noncanonical partner Sp1 upregulates GEF-H1. Our data reveal the existence of a positive feedback cycle that enhances Rho signaling through control of both GEF-H1 activation and expression. This feedback loop may play an important role in organ fibrosis.
Collapse
Affiliation(s)
- Shruthi Venugopal
- Keenan Research Centre for Biomedical Science of the St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
| | - Qinghong Dan
- Keenan Research Centre for Biomedical Science of the St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
| | - Veroni S Sri Theivakadadcham
- Keenan Research Centre for Biomedical Science of the St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
| | - Brian Wu
- Keenan Research Centre for Biomedical Science of the St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
| | - Michael Kofler
- Keenan Research Centre for Biomedical Science of the St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
| | - Matthew D Layne
- Department of Biochemistry & Cell Biology, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, United States
| | - Kim A Connelly
- Keenan Research Centre for Biomedical Science of the St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
| | - Mark F Rzepka
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Mark K Friedberg
- Division of Cardiology, Labatt Family Heart Center Toronto, Hospital for Sick Children, Toronto, Ontario, Canada
- Translational Medicine, Hospital for Sick Children Research Institute and University of Toronto, Toronto, Ontario, Canada
| | - András Kapus
- Keenan Research Centre for Biomedical Science of the St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
- Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Katalin Szászi
- Keenan Research Centre for Biomedical Science of the St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
- Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
5
|
Wang Y, Geng X, Sun X, Cui H, Guo Z, Chu D, Li J, Li Z. Celastrol alleviates subconjunctival fibrosis induced by silicone implants mimicking glaucoma surgery. Eur J Pharm Biopharm 2024; 201:114352. [PMID: 38851459 DOI: 10.1016/j.ejpb.2024.114352] [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: 02/05/2024] [Revised: 05/29/2024] [Accepted: 06/06/2024] [Indexed: 06/10/2024]
Abstract
Subconjunctival fibrosis is critical to the outcomes of several ophthalmic conditions or procedures, such as glaucoma filtering surgery. This study aimed to investigate the anti-fibrotic effect of celastrol on subconjunctival fibrosis and to further reveal the underlying mechanisms. We used celastrol-loaded nanomicelles hydrogel hybrid as a sustained-release drug. A rabbit model of subconjunctival fibrosis following silicone implantation was used for in vivo study, and TGF-β1-induced human pterygium fibroblast (HPF) activation as an in vitro model. The effects of celastrol on inhibiting TGF-β1-induced migration and proliferation of HPFs were evaluated by scratch wound assay and CCK-8, respectively. Immunofluorescence and western blotting were used to examine the effect of celastrol on the expression of α-SMA, collagen I, fibronectin, and the targets of the Hippo signaling pathway. We found that in vivo celastrol treatment reduced the expression of YAP and TAZ in subconjunctival tissue. Moreover, celastrol alleviated collagen deposition and subconjunctival fibrosis at 8 weeks. No obvious tissue toxicity was observed in the rabbit models. Mechanistically, celastrol significantly inhibited TGF-β1-induced proliferation and migration of HPFs. Pretreatment of HPFs with celastrol also suppressed the TGF-β1-induced protein expression of α-SMA, collagen I, fibronectin, TGF-βRII, phosphorylated Smad2/3, YAP, TAZ, and TEAD1. In conclusion, celastrol effectively prevented subconjunctival fibrosis through inhibiting TGF-β1/Smad2/3-YAP/TAZ pathway. Celastrol could serve as a promising therapy for subconjunctival fibrosis.
Collapse
Affiliation(s)
- Yiwei Wang
- Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou 450003, China
| | - Xingchen Geng
- Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou 450003, China
| | - Xue Sun
- Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou 450003, China
| | - Haohao Cui
- Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou 450003, China
| | - Zhihua Guo
- Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou 450003, China
| | - Dandan Chu
- Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou 450003, China
| | - Jingguo Li
- Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou 450003, China.
| | - Zhanrong Li
- Henan Eye Hospital, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou 450003, China.
| |
Collapse
|
6
|
Mohamed B, Ghareib SA, Alsemeh AE, El-Sayed SS. Telmisartan ameliorates nephropathy and restores the hippo pathway in rats with metabolic syndrome. Eur J Pharmacol 2024; 973:176605. [PMID: 38653362 DOI: 10.1016/j.ejphar.2024.176605] [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: 11/05/2023] [Revised: 04/19/2024] [Accepted: 04/19/2024] [Indexed: 04/25/2024]
Abstract
The main objective of this study was to determine if the telmisartan-ameliorative effects of metabolic syndrome (MetS)-evoked nephropathy are attributed to the Hippo pathway. A secondary objective was to investigate the potential of vitamin D3 to enhance telmisartan-favourable effects. A diet composed of 24% fat and 3% salt, along with drinking water containing 10% fructose, was administered for 12 weeks to induce MetS. MetS-rats were given telmisartan (5 mg/kg/day), vitamin D3 (10 μg/kg/day) or both by gavage, starting in the sixth week of experimental diet administration. Assessments performed at closure included renal function, histological examination, catalase, malondialdehyde (MDA), nuclear factor kappa-B (NF-κB), interleukin-6 (IL-6), peroxisome proliferator-activated receptor-γ (PPAR-γ), phosphatase and tensin homolog (PTEN), and transforming growth factor-β (TGF-β). Matrix metalloproteinase-9 (MMP-9) immunostaining was conducted. The expression of the Hippo pathway components, as well as that of angiotensin II type 1 and type 2 (AT1 and AT2), receptors was evaluated. Telmisartan attenuated MetS-evoked nephropathy, as demonstrated by improvement of renal function and histological features, enhancement of catalase, reduction of MDA, inflammation (NF-κB, IL-6), and renal fibrosis (increased PPAR-γ and PTEN and reduced MMP-9 and TGF-β). Telmisartan downregulated AT1-receptor, upregulated AT2-receptor and restored the Hippo pathway. Vitamin D3 replicated most of the telmisartan-elicited effects and enhanced the antifibrotic actions of telmisartan. The alleviative effects of telmisartan on MetS-evoked nephropathy may be related to the restoration of the Hippo pathway. The combination of vitamin D3 and telmisartan exerted more favourable effects on metabolic and nephropathic biomarkers compared with either one administered alone.
Collapse
Affiliation(s)
- Badria Mohamed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt.
| | - Salah A Ghareib
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt.
| | - Amira Ebrahim Alsemeh
- Department of Human Anatomy and Embryology, Faculty of Medicine, Zagazig University, Zagazig, 44519, Egypt.
| | - Shaimaa S El-Sayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt.
| |
Collapse
|
7
|
Bankell E, Liu L, van der Horst J, Rippe C, Jepps TA, Nilsson BO, Swärd K. Suppression of smooth muscle cell inflammation by myocardin-related transcription factors involves inactivation of TANK-binding kinase 1. Sci Rep 2024; 14:13321. [PMID: 38858497 PMCID: PMC11164896 DOI: 10.1038/s41598-024-63901-3] [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: 01/11/2024] [Accepted: 06/03/2024] [Indexed: 06/12/2024] Open
Abstract
Myocardin-related transcription factors (MRTFs: myocardin/MYOCD, MRTF-A/MRTFA, and MRTF-B/MRTFB) suppress production of pro-inflammatory cytokines and chemokines in human smooth muscle cells (SMCs) through sequestration of RelA in the NF-κB complex, but additional mechanisms are likely involved. The cGAS-STING pathway is activated by double-stranded DNA in the cytosolic compartment and acts through TANK-binding kinase 1 (TBK1) to spark inflammation. The present study tested if MRTFs suppress inflammation also by targeting cGAS-STING signaling. Interrogation of a transcriptomic dataset where myocardin was overexpressed using a panel of 56 cGAS-STING cytokines showed the panel to be repressed. Moreover, MYOCD, MRTFA, and SRF associated negatively with the panel in human arteries. RT-qPCR in human bronchial SMCs showed that all MRTFs reduced pro-inflammatory cytokines on the panel. MRTFs diminished phosphorylation of TBK1, while STING phosphorylation was marginally affected. The TBK1 inhibitor amlexanox, but not the STING inhibitor H-151, reduced the anti-inflammatory effect of MRTF-A. Co-immunoprecipitation and proximity ligation assays supported binding between MRTF-A and TBK1 in SMCs. MRTFs thus appear to suppress cellular inflammation in part by acting on the kinase TBK1. This may defend SMCs against pro-inflammatory insults in disease.
Collapse
Affiliation(s)
- Elisabeth Bankell
- Cellular Biomechanics/Vascular Physiology, Department of Experimental Medical Science, BMC D12, Lund University, 22184, Lund, Sweden
| | - Li Liu
- Cellular Biomechanics/Vascular Physiology, Department of Experimental Medical Science, BMC D12, Lund University, 22184, Lund, Sweden
- Department of Urology, Qingyuan Hospital Affiliated to Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Jennifer van der Horst
- Vascular Biology Group, Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen N, Denmark
| | - Catarina Rippe
- Cellular Biomechanics/Vascular Physiology, Department of Experimental Medical Science, BMC D12, Lund University, 22184, Lund, Sweden
| | - Thomas A Jepps
- Vascular Biology Group, Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen N, Denmark
| | - Bengt-Olof Nilsson
- Cellular Biomechanics/Vascular Physiology, Department of Experimental Medical Science, BMC D12, Lund University, 22184, Lund, Sweden
| | - Karl Swärd
- Cellular Biomechanics/Vascular Physiology, Department of Experimental Medical Science, BMC D12, Lund University, 22184, Lund, Sweden.
| |
Collapse
|
8
|
Lichner Z, Ding M, Khare T, Dan Q, Benitez R, Praszner M, Song X, Saleeb R, Hinz B, Pei Y, Szászi K, Kapus A. Myocardin-Related Transcription Factor Mediates Epithelial Fibrogenesis in Polycystic Kidney Disease. Cells 2024; 13:984. [PMID: 38891116 PMCID: PMC11172104 DOI: 10.3390/cells13110984] [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: 04/19/2024] [Revised: 05/21/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
Polycystic kidney disease (PKD) is characterized by extensive cyst formation and progressive fibrosis. However, the molecular mechanisms whereby the loss/loss-of-function of Polycystin 1 or 2 (PC1/2) provokes fibrosis are largely unknown. The small GTPase RhoA has been recently implicated in cystogenesis, and we identified the RhoA/cytoskeleton/myocardin-related transcription factor (MRTF) pathway as an emerging mediator of epithelium-induced fibrogenesis. Therefore, we hypothesized that MRTF is activated by PC1/2 loss and plays a critical role in the fibrogenic reprogramming of the epithelium. The loss of PC1 or PC2, induced by siRNA in vitro, activated RhoA and caused cytoskeletal remodeling and robust nuclear MRTF translocation and overexpression. These phenomena were also manifested in PKD1 (RC/RC) and PKD2 (WS25/-) mice, with MRTF translocation and overexpression occurring predominantly in dilated tubules and the cyst-lining epithelium, respectively. In epithelial cells, a large cohort of PC1/PC2 downregulation-induced genes was MRTF-dependent, including cytoskeletal, integrin-related, and matricellular/fibrogenic proteins. Epithelial MRTF was necessary for the paracrine priming of the fibroblast-myofibroblast transition. Thus, MRTF acts as a prime inducer of epithelial fibrogenesis in PKD. We propose that RhoA is a common upstream inducer of both histological hallmarks of PKD: cystogenesis and fibrosis.
Collapse
Affiliation(s)
- Zsuzsanna Lichner
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada; (Z.L.); (T.K.); (R.S.); (K.S.)
| | - Mei Ding
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada; (Z.L.); (T.K.); (R.S.); (K.S.)
| | - Tarang Khare
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada; (Z.L.); (T.K.); (R.S.); (K.S.)
- Enrich Bioscience, Toronto, ON M5B 1T8, Canada
| | - Qinghong Dan
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada; (Z.L.); (T.K.); (R.S.); (K.S.)
| | - Raquel Benitez
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada; (Z.L.); (T.K.); (R.S.); (K.S.)
| | - Mercédesz Praszner
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada; (Z.L.); (T.K.); (R.S.); (K.S.)
| | - Xuewen Song
- Division of Nephrology, University Health Network, Toronto, ON M5G 2C4, Canada
| | - Rola Saleeb
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada; (Z.L.); (T.K.); (R.S.); (K.S.)
- Department of Laboratory Medicine and Pathobiology, Temerty School of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Boris Hinz
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada; (Z.L.); (T.K.); (R.S.); (K.S.)
- Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada
| | - York Pei
- Division of Nephrology, University Health Network, Toronto, ON M5G 2C4, Canada
| | - Katalin Szászi
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada; (Z.L.); (T.K.); (R.S.); (K.S.)
- Department of Laboratory Medicine and Pathobiology, Temerty School of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of Surgery, University of Toronto, Toronto, ON M5T 1P5, Canada
| | - András Kapus
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada; (Z.L.); (T.K.); (R.S.); (K.S.)
- Department of Surgery, University of Toronto, Toronto, ON M5T 1P5, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
| |
Collapse
|
9
|
Zhang Y, Ren Y, Li X, Li M, Fu M, Zhou W, Yu Y, Xiong Y. A review on decoding the roles of YAP/TAZ signaling pathway in cardiovascular diseases: Bridging molecular mechanisms to therapeutic insights. Int J Biol Macromol 2024; 271:132473. [PMID: 38795886 DOI: 10.1016/j.ijbiomac.2024.132473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 05/02/2024] [Accepted: 05/15/2024] [Indexed: 05/28/2024]
Abstract
Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) serve as transcriptional co-activators that dynamically shuttle between the cytoplasm and nucleus, resulting in either the suppression or enhancement of their downstream gene expression. Recent emerging evidence demonstrates that YAP/TAZ is strongly implicated in the pathophysiological processes that contribute to cardiovascular diseases (CVDs). In the cardiovascular system, YAP/TAZ is involved in the orchestration of a range of biological processes such as oxidative stress, inflammation, proliferation, and autophagy. Furthermore, YAP/TAZ has been revealed to be closely associated with the initiation and development of various cardiovascular diseases, including atherosclerosis, pulmonary hypertension, myocardial fibrosis, cardiac hypertrophy, and cardiomyopathy. In this review, we delve into recent studies surrounding YAP and TAZ, along with delineating their roles in contributing to the pathogenesis of CVDs with a link to various physiological processes in the cardiovascular system. Additionally, we highlight the current potential drugs targeting YAP/TAZ for CVDs therapy and discuss their challenges for translational application. Overall, this review may offer novel insights for understanding and treating cardiovascular disorders.
Collapse
Affiliation(s)
- Yan Zhang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an 710069, Shaanxi, PR China
| | - Yuanyuan Ren
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an 710069, Shaanxi, PR China
| | - Xiaofang Li
- Department of Gastroenterology, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Xi'an, Shaanxi 710018, PR China
| | - Man Li
- Department of Endocrinology, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Xi'an, Shaanxi 710018, PR China
| | - Mingdi Fu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an 710069, Shaanxi, PR China
| | - Wenjing Zhou
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an 710069, Shaanxi, PR China
| | - Yi Yu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an 710069, Shaanxi, PR China.
| | - Yuyan Xiong
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an 710069, Shaanxi, PR China; Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, the Affiliated Hospital of Northwest University, 710018 Xi'an, Shaanxi, PR China.
| |
Collapse
|
10
|
Singh B, Cui K, Eisa-Beygi S, Zhu B, Cowan DB, Shi J, Wang DZ, Liu Z, Bischoff J, Chen H. Elucidating the crosstalk between endothelial-to-mesenchymal transition (EndoMT) and endothelial autophagy in the pathogenesis of atherosclerosis. Vascul Pharmacol 2024; 155:107368. [PMID: 38548093 PMCID: PMC11303600 DOI: 10.1016/j.vph.2024.107368] [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: 01/17/2024] [Revised: 03/07/2024] [Accepted: 03/25/2024] [Indexed: 04/26/2024]
Abstract
Atherosclerosis, a chronic systemic inflammatory condition, is implicated in most cardiovascular ischemic events. The pathophysiology of atherosclerosis involves various cell types and associated processes, including endothelial cell activation, monocyte recruitment, smooth muscle cell migration, involvement of macrophages and foam cells, and instability of the extracellular matrix. The process of endothelial-to-mesenchymal transition (EndoMT) has recently emerged as a pivotal process in mediating vascular inflammation associated with atherosclerosis. This transition occurs gradually, with a significant portion of endothelial cells adopting an intermediate state, characterized by a partial loss of endothelial-specific gene expression and the acquisition of "mesenchymal" traits. Consequently, this shift disrupts endothelial cell junctions, increases vascular permeability, and exacerbates inflammation, creating a self-perpetuating cycle that drives atherosclerotic progression. While endothelial cell dysfunction initiates the development of atherosclerosis, autophagy, a cellular catabolic process designed to safeguard cells by recycling intracellular molecules, is believed to exert a significant role in plaque development. Identifying the pathological mechanisms and molecular mediators of EndoMT underpinning endothelial autophagy, may be of clinical relevance. Here, we offer new insights into the underlying biology of atherosclerosis and present potential molecular mechanisms of atherosclerotic resistance and highlight potential therapeutic targets.
Collapse
Affiliation(s)
- Bandana Singh
- Vascular Biology Program, Department of Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Kui Cui
- Vascular Biology Program, Department of Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Shahram Eisa-Beygi
- Vascular Biology Program, Department of Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Bo Zhu
- Vascular Biology Program, Department of Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Douglas B Cowan
- Vascular Biology Program, Department of Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Jinjun Shi
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Da-Zhi Wang
- Center for Regenerative Medicine, University of South Florida Health Heart Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Zhenguo Liu
- Division of Cardiovascular Medicine, Department of Medicine, University of Missouri School of Medicine, Columbia, MO, USA
| | - Joyce Bischoff
- Vascular Biology Program, Department of Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Hong Chen
- Vascular Biology Program, Department of Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA.
| |
Collapse
|
11
|
Leong E, Al-Bitar H, Marshall JS, Bezuhly M. Ketotifen directly modifies the fibrotic response of human skin fibroblasts. Sci Rep 2024; 14:7076. [PMID: 38528089 DOI: 10.1038/s41598-024-57776-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 03/21/2024] [Indexed: 03/27/2024] Open
Abstract
Fibrosis is a destructive, end-stage disease process. In the skin, it is associated with systemic sclerosis and scarring with considerable health burden. Ketotifen is a clinical antihistamine and mast cell stabilizer. Studies have demonstrated mast cell-dependent anti-fibrotic effects of ketotifen but direct effects on fibroblasts have not been determined. Human dermal fibroblasts were treated with pro-fibrotic transforming growth factor-β1 (TGFβ) followed by ketotifen or control treatments to determine direct effects on fibrotic fibroblasts. Ketotifen impaired TGFβ-induced α-smooth muscle actin gene and protein responses and decreased cytoskeletal- and contractility-associated gene responses associated with fibrosis. Ketotifen reduced Yes-associated protein phosphorylation, transcriptional coactivator with PDZ binding motif transcript and protein levels, and phosphorylation of protein kinase B. In a fibroblast-populated collagen gel contraction assay, ketotifen reduced the contractile activity of TGFβ-activated fibroblasts. In a murine model of bleomycin-induced skin fibrosis, collagen density and dermal thickness were significantly decreased in ketotifen-treated mice supporting in vitro findings. These results support a novel, direct anti-fibrotic activity of ketotifen, reducing pro-fibrotic phenotypic changes in fibroblasts and reducing collagen fibres in fibrotic mouse skin. Together, these findings suggest novel therapeutic potential and a novel mechanism of action for ketotifen in the context of fibrosis.
Collapse
Affiliation(s)
- Edwin Leong
- Department of Pathology, Dalhousie University, 5850 College Street, Room 7-C, PO BOX 15000, Halifax, NS, B3H 4R2, Canada
| | - Haya Al-Bitar
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Canada
| | - Jean S Marshall
- Department of Pathology, Dalhousie University, 5850 College Street, Room 7-C, PO BOX 15000, Halifax, NS, B3H 4R2, Canada.
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Canada.
- Beatrice Hunter Cancer Research Institute, Halifax, Canada.
| | - Michael Bezuhly
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Canada.
- Beatrice Hunter Cancer Research Institute, Halifax, Canada.
- Division of Plastic Surgery, Izaak Walton Killam Health Centre, 5850/5980 University Avenue, PO Box 9700, Halifax, NS, B3K 6R8, Canada.
- Department of Surgery, Dalhousie University, Halifax, Canada.
| |
Collapse
|
12
|
Ríos-López DG, Tecalco-Cruz AC, Martínez-Pastor D, Sosa-Garrocho M, Tapia-Urzúa G, Aranda-López Y, Ortega-Domínguez B, Recillas-Targa F, Vázquez-Victorio G, Macías-Silva M. TGF-β/SMAD canonical pathway induces the expression of transcriptional cofactor TAZ in liver cancer cells. Heliyon 2023; 9:e21519. [PMID: 38027697 PMCID: PMC10660035 DOI: 10.1016/j.heliyon.2023.e21519] [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: 04/06/2023] [Revised: 10/20/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
The TGF-β and Hippo pathways are critical for liver size control, regeneration, and cancer progression. The transcriptional cofactor TAZ, also named WWTR1, is a downstream effector of Hippo pathway and plays a key role in the maintenance of liver physiological functions. However, the up-regulation of TAZ expression has been associated with liver cancer progression. Recent evidence shows crosstalk of TGF-β and Hippo pathways, since TGF-β modulates TAZ expression through different mechanisms in a cellular context-dependent manner but supposedly independent of SMADs. Here, we evaluate the molecular interplay between TGF-β pathway and TAZ expression and observe that TGF-β induces TAZ expression through SMAD canonical pathway in liver cancer HepG2 cells. Therefore, TAZ cofactor is a primary target of TGF-β/SMAD-signaling, one of the pathways altered in liver cancer.
Collapse
Affiliation(s)
- Diana G. Ríos-López
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Angeles C. Tecalco-Cruz
- Programa en Ciencias Genómicas, Universidad Autónoma de La Ciudad de México, Ciudad de México 03100, Mexico
| | - David Martínez-Pastor
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Marcela Sosa-Garrocho
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Gustavo Tapia-Urzúa
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Yuli Aranda-López
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Bibiana Ortega-Domínguez
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Félix Recillas-Targa
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Genaro Vázquez-Victorio
- Facultad de Ciencias, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Marina Macías-Silva
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| |
Collapse
|
13
|
Small GW, Akhtari FS, Green AJ, Havener TM, Sikes M, Quintanhila J, Gonzalez RD, Reif DM, Motsinger-Reif AA, McLeod HL, Wiltshire T. Pharmacogenomic Analyses Implicate B Cell Developmental Status and MKL1 as Determinants of Sensitivity toward Anti-CD20 Monoclonal Antibody Therapy. Cells 2023; 12:1574. [PMID: 37371044 DOI: 10.3390/cells12121574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 06/02/2023] [Accepted: 06/04/2023] [Indexed: 06/29/2023] Open
Abstract
Monoclonal antibody (mAb) therapy directed against CD20 is an important tool in the treatment of B cell disorders. However, variable patient response and acquired resistance remain important clinical challenges. To identify genetic factors that may influence sensitivity to treatment, the cytotoxic activity of three CD20 mAbs: rituximab; ofatumumab; and obinutuzumab, were screened in high-throughput assays using 680 ethnically diverse lymphoblastoid cell lines (LCLs) followed by a pharmacogenomic assessment. GWAS analysis identified several novel gene candidates. The most significant SNP, rs58600101, in the gene MKL1 displayed ethnic stratification, with the variant being significantly more prevalent in the African cohort and resulting in reduced transcript levels as measured by qPCR. Functional validation of MKL1 by shRNA-mediated knockdown of MKL1 resulted in a more resistant phenotype. Gene expression analysis identified the developmentally associated TGFB1I1 as the most significant gene associated with sensitivity. qPCR among a panel of sensitive and resistant LCLs revealed immunoglobulin class-switching as well as differences in the expression of B cell activation markers. Flow cytometry showed heterogeneity within some cell lines relative to surface Ig isotype with a shift to more IgG+ cells among the resistant lines. Pretreatment with prednisolone could partly reverse the resistant phenotype. Results suggest that the efficacy of anti-CD20 mAb therapy may be influenced by B cell developmental status as well as polymorphism in the MKL1 gene. A clinical benefit may be achieved by pretreatment with corticosteroids such as prednisolone followed by mAb therapy.
Collapse
Affiliation(s)
- George W Small
- Pharmacotherapy and Experimental Therapeutics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Farida S Akhtari
- Biostatistics and Computational Biology Branch, Division of Intramural Research, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Adrian J Green
- Department of Biological Sciences, Bioinformatics Research Center, North Carolina State University, Raleigh, NC 27695, USA
| | - Tammy M Havener
- Structural Genomics Consortium and Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Michael Sikes
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, USA
| | | | - Ricardo D Gonzalez
- Pharmacotherapy and Experimental Therapeutics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - David M Reif
- Predictive Toxicology Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Alison A Motsinger-Reif
- Biostatistics and Computational Biology Branch, Division of Intramural Research, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Howard L McLeod
- Center for Precision Medicine and Functional Genomics, Utah Tech University, 225 South University Ave, St. George, UT 84770, USA
| | - Tim Wiltshire
- Pharmacotherapy and Experimental Therapeutics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Center for Pharmacogenomics and Individualized Therapy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
14
|
Messina B, Lo Sardo F, Scalera S, Memeo L, Colarossi C, Mare M, Blandino G, Ciliberto G, Maugeri-Saccà M, Bon G. Hippo pathway dysregulation in gastric cancer: from Helicobacter pylori infection to tumor promotion and progression. Cell Death Dis 2023; 14:21. [PMID: 36635265 PMCID: PMC9837097 DOI: 10.1038/s41419-023-05568-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/27/2022] [Accepted: 01/06/2023] [Indexed: 01/13/2023]
Abstract
The Hippo pathway plays a critical role for balancing proliferation and differentiation, thus regulating tissue homeostasis. The pathway acts through a kinase cascade whose final effectors are the Yes-associated protein (YAP) and its paralog transcriptional co‑activator with PDZ‑binding motif (TAZ). In response to a variety of upstream signals, YAP and TAZ activate a transcriptional program that modulates cellular proliferation, tissue repair after injury, stem cell fate decision, and cytoskeletal reorganization. Hippo pathway signaling is often dysregulated in gastric cancer and in Helicobacter pylori-induced infection, suggesting a putative role of its deregulation since the early stages of the disease. In this review, we summarize the architecture and regulation of the Hippo pathway and discuss how its dysregulation fuels the onset and progression of gastric cancer. In this setting, we also focus on the crosstalk between Hippo and other established oncogenic signaling pathways. Lastly, we provide insights into the therapeutic approaches targeting aberrant YAP/TAZ activation and discuss the related clinical perspectives and challenges.
Collapse
Affiliation(s)
- Beatrice Messina
- Clinical Trial Center, Biostatistics and Bioinformatics Unit, Department of Research, Diagnosis and Innovative Technologies, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Federica Lo Sardo
- Oncogenomic and Epigenetic Unit, Department of Research, Diagnosis and Innovative Technologies, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Stefano Scalera
- SAFU Laboratory, Department of Research, Advanced Diagnostic, and Technological Innovation, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Lorenzo Memeo
- Pathology Unit, Mediterranean Institute of Oncology, Viagrande, Italy
| | | | - Marzia Mare
- Medical Oncology Unit, Mediterranean Institute of Oncology, Viagrande, Italy
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences, University of Messina, Messina, Italy
| | - Giovanni Blandino
- Oncogenomic and Epigenetic Unit, Department of Research, Diagnosis and Innovative Technologies, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Gennaro Ciliberto
- Scientific Directorate, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Marcello Maugeri-Saccà
- Clinical Trial Center, Biostatistics and Bioinformatics Unit, Department of Research, Diagnosis and Innovative Technologies, IRCCS Regina Elena National Cancer Institute, Rome, Italy
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Giulia Bon
- Cellular Network and Molecular Therapeutic Target Unit, Department of Research, Diagnosis and Innovative Technologies, IRCCS Regina Elena National Cancer Institute, Rome, Italy.
| |
Collapse
|
15
|
Schuster R, Younesi F, Ezzo M, Hinz B. The Role of Myofibroblasts in Physiological and Pathological Tissue Repair. Cold Spring Harb Perspect Biol 2023; 15:cshperspect.a041231. [PMID: 36123034 PMCID: PMC9808581 DOI: 10.1101/cshperspect.a041231] [Citation(s) in RCA: 44] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Myofibroblasts are the construction workers of wound healing and repair damaged tissues by producing and organizing collagen/extracellular matrix (ECM) into scar tissue. Scar tissue effectively and quickly restores the mechanical integrity of lost tissue architecture but comes at the price of lost tissue functionality. Fibrotic diseases caused by excessive or persistent myofibroblast activity can lead to organ failure. This review defines myofibroblast terminology, phenotypic characteristics, and functions. We will focus on the central role of the cell, ECM, and tissue mechanics in regulating tissue repair by controlling myofibroblast action. Additionally, we will discuss how therapies based on mechanical intervention potentially ameliorate wound healing outcomes. Although myofibroblast physiology and pathology affect all organs, we will emphasize cutaneous wound healing and hypertrophic scarring as paradigms for normal tissue repair versus fibrosis. A central message of this review is that myofibroblasts can be activated from multiple cell sources, varying with local environment and type of injury, to either restore tissue integrity and organ function or create an inappropriate mechanical environment.
Collapse
Affiliation(s)
- Ronen Schuster
- Faculty of Dentistry, University of Toronto, Toronto, M5S 3E2 Ontario, Canada
| | - Fereshteh Younesi
- Faculty of Dentistry, University of Toronto, Toronto, M5S 3E2 Ontario, Canada.,Laboratory of Tissue Repair and Regeneration, Keenan Research Centre for Biomedical Science of the St. Michael's Hospital, Toronto, Ontario M5B 1T8, Canada
| | - Maya Ezzo
- Faculty of Dentistry, University of Toronto, Toronto, M5S 3E2 Ontario, Canada.,Laboratory of Tissue Repair and Regeneration, Keenan Research Centre for Biomedical Science of the St. Michael's Hospital, Toronto, Ontario M5B 1T8, Canada
| | - Boris Hinz
- Faculty of Dentistry, University of Toronto, Toronto, M5S 3E2 Ontario, Canada.,Laboratory of Tissue Repair and Regeneration, Keenan Research Centre for Biomedical Science of the St. Michael's Hospital, Toronto, Ontario M5B 1T8, Canada
| |
Collapse
|
16
|
Transcriptional Regulation of Siglec-15 by ETS-1 and ETS-2 in Hepatocellular Carcinoma Cells. Int J Mol Sci 2023; 24:ijms24010792. [PMID: 36614238 PMCID: PMC9821606 DOI: 10.3390/ijms24010792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 12/06/2022] [Accepted: 12/07/2022] [Indexed: 01/04/2023] Open
Abstract
Sialic acid-binding immunoglobulin-like lectin 15 (Siglec-15) has been identified as a crucial immune suppressor in human cancers, comparable to programmed cell death 1 ligand (PD-L1). However, the regulatory mechanisms underlying its transcriptional upregulation in human cancers remain largely unknown. Here, we show that the transcription factors ETS-1 and ETS-2 bound to the Siglec-15 promoter to enhance transcription and expression of Siglec-15 in hepatocellular carcinoma (HCC) cells and that transforming growth factor β-1 (TGF-β1) upregulated the expression of ETS-1 and ETS-2 and facilitated the binding of ETS-1 and ETS-2 to the Siglec-15 promoter. We further demonstrate that TGF-β1 activated the Ras/C-Raf/MEK/ERK1/2 signaling pathway, leading to phosphorylation of ETS-1 and ETS-2, which consequently upregulates the transcription and expression of Siglec-15. Our study defines a detailed molecular profile of how Siglec-15 is transcriptionally regulated which may offer significant opportunity for therapeutic intervention on HCC immunotherapy.
Collapse
|
17
|
Hayashi K, Labios RJ, Morita T, Ashimori A, Aoki R, Mikuni M, Kimura K. Significance of the p38MAPK-CRP2 axis in myofibroblastic phenotypic transition. Cell Struct Funct 2023; 48:199-210. [PMID: 37899269 PMCID: PMC11496777 DOI: 10.1247/csf.23060] [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: 08/23/2023] [Accepted: 09/20/2023] [Indexed: 10/31/2023] Open
Abstract
We have recently demonstrated that a LIM domain protein, cysteine and glycine-rich protein 2 (CSRP2 [CRP2]), plays a vital role in the functional expression of myofibroblasts and cancer-associated fibroblasts. CRP2 binds directly to myocardin-related transcription factors (MRTF [MRTF-A or MRTF-B]) and serum response factor (SRF) to stabilize the MRTF/SRF/CArG-box complex, leading to the expression of smooth muscle cell (SMC) genes such as α-smooth muscle actin (α-SMA) and collagens. These are the marker genes for myofibroblasts. Here, we show that the adhesion of cultured human skin fibroblasts (HSFs) to collagen reduces the myofibroblastic features. HSF adhesion to collagen suppresses the expression of CRP2 and CSRP2-binding protein (CSRP2BP [CRP2BP]) and reduces the expression of SMC genes. Although CRP2BP is known as an epigenetic factor, we find that CRP2BP also acts as an adaptor protein to enhance the function of CRP2 mentioned above. This CRP2BP function does not depend on its histone acetyltransferase activity. We also addressed the molecular mechanism of the reduced myofibroblastic features of HSFs on collagen. HSF adhesion to collagen inhibits the p38MAPK-mediated pathway, and reducing the p38MAPK activity decreases the expression of CRP2 and SMC genes. Thus, the activation of p38MAPK is critical for the myofibroblastic features. We also show evidence that CRP2 plays a role in the myofibroblastic transition of retinal pigment epithelial cells (RPEs). Like HSFs, such phenotypic modulation of RPEs depends on the p38MAPK pathway.Key words: CRP2, p38MAPK, MRTF, myofibroblasts, retinal pigment epithelial cells.
Collapse
Affiliation(s)
- Ken’ichiro Hayashi
- Department of RNA Biology and Neuroscience, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
- Department of Ophthalmology, Yamaguchi University Graduate School of Medicine, Minami-Kogushi 1-1-1, Ube, Yamaguchi 755-8505, Japan
| | - Reuben Jacob Labios
- Department of RNA Biology and Neuroscience, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Tsuyoshi Morita
- Department of Biology, Wakayama Medical University School of Medicine, 580 Mikazura, Wakayama 641-0011, Japan
| | - Atsushige Ashimori
- Department of Ophthalmology, Yamaguchi University Graduate School of Medicine, Minami-Kogushi 1-1-1, Ube, Yamaguchi 755-8505, Japan
| | - Ren Aoki
- Department of Ophthalmology, Yamaguchi University Graduate School of Medicine, Minami-Kogushi 1-1-1, Ube, Yamaguchi 755-8505, Japan
| | - Masanori Mikuni
- Department of Ophthalmology, Yamaguchi University Graduate School of Medicine, Minami-Kogushi 1-1-1, Ube, Yamaguchi 755-8505, Japan
| | - Kazuhiro Kimura
- Department of Ophthalmology, Yamaguchi University Graduate School of Medicine, Minami-Kogushi 1-1-1, Ube, Yamaguchi 755-8505, Japan
| |
Collapse
|
18
|
Bhavnagari H, Raval A, Shah F. Deciphering Potential Role of Hippo Signaling Pathway in Breast Cancer: A Comprehensive Review. Curr Pharm Des 2023; 29:3505-3518. [PMID: 38141194 DOI: 10.2174/0113816128274418231215054210] [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: 07/28/2023] [Revised: 11/16/2023] [Accepted: 11/23/2023] [Indexed: 12/25/2023]
Abstract
Breast cancer is a heterogeneous disease and a leading malignancy around the world. It is a vital cause of untimely mortality among women. Drug resistance is the major challenge for effective cancer therapeutics. In contrast, cancer stem cells (CSCs) are one of the reasons for drug resistance, tumor progression, and metastasis. The small population of CSCs present in each tumor has the ability of self-renewal, differentiation, and tumorigenicity. CSCs are often identified and enriched using a variety of cell surface markers (CD44, CD24, CD133, ABCG2, CD49f, LGR5, SSEA-3, CD70) that exert their functions by different regulatory networks, i.e., Notch, Wnt/β-catenin, hedgehog (Hh), and Hippo signaling pathways. Particularly the Hippo signaling pathway is the emerging and very less explored cancer stem cell pathway. Here, in this review, the Hippo signaling molecules are elaborated with respect to their ability of stemness as epigenetic modulators and how these molecules can be targeted for better cancer treatment and to overcome drug resistance.
Collapse
Affiliation(s)
- Hunayna Bhavnagari
- Molecular Diagnostic and Research Lab-3, Department of Cancer Biology, The Gujarat Cancer and Research Institute, Ahmedabad, Gujarat, India
| | - Apexa Raval
- Molecular Diagnostic and Research Lab-3, Department of Cancer Biology, The Gujarat Cancer and Research Institute, Ahmedabad, Gujarat, India
| | - Franky Shah
- Molecular Diagnostic and Research Lab-3, Department of Cancer Biology, The Gujarat Cancer and Research Institute, Ahmedabad, Gujarat, India
| |
Collapse
|
19
|
Garcia K, Gingras AC, Harvey KF, Tanas MR. TAZ/YAP fusion proteins: mechanistic insights and therapeutic opportunities. Trends Cancer 2022; 8:1033-1045. [PMID: 36096997 PMCID: PMC9671862 DOI: 10.1016/j.trecan.2022.08.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 08/07/2022] [Accepted: 08/09/2022] [Indexed: 12/24/2022]
Abstract
The Hippo pathway is dysregulated in many different cancers, but point mutations in the pathway are rare. Transcriptional co-activator with PDZ-binding motif (TAZ) and Yes-associated protein (YAP) fusion proteins have emerged in almost all major cancer types and represent the most common genetic mechanism by which the two transcriptional co-activators are activated. Given that the N termini of TAZ or YAP are fused to the C terminus of another transcriptional regulator, the resultant fusion proteins hyperactivate a TEAD transcription factor-based transcriptome. Recent advances show that the C-terminal fusion partners confer oncogenic properties to TAZ/YAP fusion proteins by recruiting epigenetic modifiers that promote a hybrid TEAD-based transcriptome. Elucidating these cooperating epigenetic complexes represents a strategy to identify new therapeutic approaches for a pathway that has been recalcitrant to medical therapy.
Collapse
Affiliation(s)
- Keith Garcia
- Department of Pathology, University of Iowa, Iowa City, IA, USA; Cancer Biology Graduate Program, University of Iowa, Iowa City, IA, USA
| | - Anne-Claude Gingras
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Kieran F Harvey
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia; Department of Anatomy and Developmental Biology, and Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Munir R Tanas
- Department of Pathology, University of Iowa, Iowa City, IA, USA; Cancer Biology Graduate Program, University of Iowa, Iowa City, IA, USA; Pathology and Laboratory Medicine, Veterans Affairs Medical Center, Iowa City, IA, USA; Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
20
|
Fu M, Hu Y, Lan T, Guan KL, Luo T, Luo M. The Hippo signalling pathway and its implications in human health and diseases. Signal Transduct Target Ther 2022; 7:376. [PMID: 36347846 PMCID: PMC9643504 DOI: 10.1038/s41392-022-01191-9] [Citation(s) in RCA: 152] [Impact Index Per Article: 76.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 09/09/2022] [Accepted: 09/09/2022] [Indexed: 11/11/2022] Open
Abstract
As an evolutionarily conserved signalling network, the Hippo pathway plays a crucial role in the regulation of numerous biological processes. Thus, substantial efforts have been made to understand the upstream signals that influence the activity of the Hippo pathway, as well as its physiological functions, such as cell proliferation and differentiation, organ growth, embryogenesis, and tissue regeneration/wound healing. However, dysregulation of the Hippo pathway can cause a variety of diseases, including cancer, eye diseases, cardiac diseases, pulmonary diseases, renal diseases, hepatic diseases, and immune dysfunction. Therefore, therapeutic strategies that target dysregulated Hippo components might be promising approaches for the treatment of a wide spectrum of diseases. Here, we review the key components and upstream signals of the Hippo pathway, as well as the critical physiological functions controlled by the Hippo pathway. Additionally, diseases associated with alterations in the Hippo pathway and potential therapies targeting Hippo components will be discussed.
Collapse
Affiliation(s)
- Minyang Fu
- Breast Disease Center, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, South of Renmin Road, 610041, Chengdu, China
| | - Yuan Hu
- Department of Pediatric Nephrology Nursing, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, 610041, Chengdu, China
| | - Tianxia Lan
- Breast Disease Center, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, South of Renmin Road, 610041, Chengdu, China
| | - Kun-Liang Guan
- Department of Pharmacology and Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Ting Luo
- Breast Disease Center, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, South of Renmin Road, 610041, Chengdu, China.
| | - Min Luo
- Breast Disease Center, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, South of Renmin Road, 610041, Chengdu, China.
| |
Collapse
|
21
|
Zeitlmayr S, Zierler S, Staab-Weijnitz CA, Dietrich A, Geiger F, Horgen FD, Gudermann T, Breit A. TRPM7 restrains plasmin activity and promotes transforming growth factor-β1 signaling in primary human lung fibroblasts. Arch Toxicol 2022; 96:2767-2783. [PMID: 35864199 PMCID: PMC9302958 DOI: 10.1007/s00204-022-03342-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 07/14/2022] [Indexed: 02/07/2023]
Abstract
Sustained exposure of the lung to various environmental or occupational toxins may eventually lead to pulmonary fibrosis, a devastating disease with no cure. Pulmonary fibrosis is characterized by excessive deposition of extracellular matrix (ECM) proteins such as fibronectin and collagens. The peptidase plasmin degrades the ECM, but protein levels of the plasmin activator inhibitor-1 (PAI-1) are increased in fibrotic lung tissue, thereby dampening plasmin activity. Transforming growth factor-β1 (TGF-β1)-induced activation of SMAD transcription factors promotes ECM deposition by enhancing collagen, fibronectin and PAI-1 levels in pulmonary fibroblasts. Hence, counteracting TGF-β1-induced signaling is a promising approach for the therapy of pulmonary fibrosis. Transient receptor potential cation channel subfamily M Member 7 (TRPM7) supports TGF-β1-promoted SMAD signaling in T-lymphocytes and the progression of fibrosis in kidney and heart. Thus, we investigated possible effects of TRPM7 on plasmin activity, ECM levels and TGF-β1 signaling in primary human pulmonary fibroblasts (pHPF). We found that two structurally unrelated TRPM7 blockers enhanced plasmin activity and reduced fibronectin or PAI-1 protein levels in pHPF under basal conditions. Further, TRPM7 blockade strongly inhibited fibronectin and collagen deposition induced by sustained TGF-β1 stimulation. In line with these data, inhibition of TRPM7 activity diminished TGF-β1-triggered phosphorylation of SMAD-2, SMAD-3/4-dependent reporter activation and PAI-1 mRNA levels. Overall, we uncover TRPM7 as a novel supporter of TGF-β1 signaling in pHPF and propose TRPM7 blockers as new candidates to control excessive ECM levels under pathophysiological conditions conducive to pulmonary fibrosis.
Collapse
Affiliation(s)
- Sarah Zeitlmayr
- Walther Straub Institute of Pharmacology and Toxicology, Medical Faculty, LMU Munich, Goethestrasse 33, 80336, Munich, Germany
| | - Susanna Zierler
- Walther Straub Institute of Pharmacology and Toxicology, Medical Faculty, LMU Munich, Goethestrasse 33, 80336, Munich, Germany.,Faculty of Medicine, Johannes Kepler University, Life Science Park, Huemerstraße 3-5, 4020, Linz, Austria
| | - Claudia A Staab-Weijnitz
- Institute of Lung Health and Immunity and Comprehensive Pneumology Center, Helmholtz Zentrum München GmbH, Member of the German Center for Lung Research, Max-Lebsche-Platz 31, 81377, Munich, Germany
| | - Alexander Dietrich
- Walther Straub Institute of Pharmacology and Toxicology, Medical Faculty, LMU Munich, Goethestrasse 33, 80336, Munich, Germany
| | - Fabienne Geiger
- Walther Straub Institute of Pharmacology and Toxicology, Medical Faculty, LMU Munich, Goethestrasse 33, 80336, Munich, Germany
| | - F David Horgen
- Department of Natural Sciences, Hawaii Pacific University, Kaneohe, HI, 96744, USA
| | - Thomas Gudermann
- Walther Straub Institute of Pharmacology and Toxicology, Medical Faculty, LMU Munich, Goethestrasse 33, 80336, Munich, Germany
| | - Andreas Breit
- Walther Straub Institute of Pharmacology and Toxicology, Medical Faculty, LMU Munich, Goethestrasse 33, 80336, Munich, Germany.
| |
Collapse
|
22
|
Zhang T, He X, Caldwell L, Goru SK, Ulloa Severino L, Tolosa MF, Misra PS, McEvoy CM, Christova T, Liu Y, Atin C, Zhang J, Hu C, Vukosa N, Chen X, Krizova A, Kirpalani A, Gregorieff A, Ni R, Chan K, Gill MK, Attisano L, Wrana JL, Yuen DA. NUAK1 promotes organ fibrosis via YAP and TGF-β/SMAD signaling. Sci Transl Med 2022; 14:eaaz4028. [PMID: 35320001 DOI: 10.1126/scitranslmed.aaz4028] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Fibrosis is a central pathway that drives progression of multiple chronic diseases, yet few safe and effective clinical antifibrotic therapies exist. In most fibrotic disorders, transforming growth factor-β (TGF-β)-driven scarring is an important pathologic feature and a key contributor to disease progression. Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) are two closely related transcription cofactors that are important for coordinating fibrogenesis after organ injury, but how they are activated in response to tissue injury has, so far, remained unclear. Here, we describe NUAK family kinase 1 (NUAK1) as a TGF-β-inducible profibrotic kinase that is up-regulated in multiple fibrotic organs in mice and humans. Mechanistically, we show that TGF-β induces a rapid increase in NUAK1 in fibroblasts. NUAK1, in turn, can promote profibrotic YAP and TGF-β/SMAD signaling, ultimately leading to organ scarring. Moreover, activated YAP and TAZ can induce further NUAK1 expression, creating a profibrotic positive feedback loop that enables persistent fibrosis. Using mouse models of kidney, lung, and liver fibrosis, we demonstrate that this fibrogenic signaling loop can be interrupted via fibroblast-specific loss of NUAK1 expression, leading to marked attenuation of fibrosis. Pharmacologic NUAK1 inhibition also reduced scarring, either when initiated immediately after injury or when initiated after fibrosis was already established. Together, our data suggest that NUAK1 plays a critical, previously unrecognized role in fibrogenesis and represents an attractive target for strategies that aim to slow fibrotic disease progression.
Collapse
Affiliation(s)
- Tianzhou Zhang
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital (Unity Health Toronto) and Department of Medicine, University of Toronto, Toronto, Ontario M5B 1T8, Canada
| | - Xiaolin He
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital (Unity Health Toronto) and Department of Medicine, University of Toronto, Toronto, Ontario M5B 1T8, Canada
| | - Lauren Caldwell
- Center for Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital and Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5G 1X5, Canada
| | - Santosh Kumar Goru
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital (Unity Health Toronto) and Department of Medicine, University of Toronto, Toronto, Ontario M5B 1T8, Canada
| | - Luisa Ulloa Severino
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital (Unity Health Toronto) and Department of Medicine, University of Toronto, Toronto, Ontario M5B 1T8, Canada
| | - Monica F Tolosa
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital (Unity Health Toronto) and Department of Medicine, University of Toronto, Toronto, Ontario M5B 1T8, Canada
| | - Paraish S Misra
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital (Unity Health Toronto) and Department of Medicine, University of Toronto, Toronto, Ontario M5B 1T8, Canada
| | - Caitríona M McEvoy
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital (Unity Health Toronto) and Department of Medicine, University of Toronto, Toronto, Ontario M5B 1T8, Canada
| | - Tania Christova
- Donnelly Centre and Department of Biochemistry, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Yong Liu
- Ontario Institute of Cancer Research, Toronto, Ontario M5G OA3, Canada
| | - Cassandra Atin
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital (Unity Health Toronto) and Department of Medicine, University of Toronto, Toronto, Ontario M5B 1T8, Canada
| | - Johnny Zhang
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital (Unity Health Toronto) and Department of Medicine, University of Toronto, Toronto, Ontario M5B 1T8, Canada
| | - Catherine Hu
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital (Unity Health Toronto) and Department of Medicine, University of Toronto, Toronto, Ontario M5B 1T8, Canada
| | - Noah Vukosa
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital (Unity Health Toronto) and Department of Medicine, University of Toronto, Toronto, Ontario M5B 1T8, Canada
| | - Xiaolan Chen
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital (Unity Health Toronto) and Department of Medicine, University of Toronto, Toronto, Ontario M5B 1T8, Canada
| | - Adriana Krizova
- Department of Laboratory Medicine and Pathobiology, School of Graduate Studies, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Anish Kirpalani
- Department of Medical Imaging, St. Michael's Hospital (Unity Health Toronto) and University of Toronto, Toronto, Ontario M5B 1W8, Canada
| | - Alex Gregorieff
- Center for Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital and Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5G 1X5, Canada
| | - Ruoyu Ni
- Center for Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital and Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5G 1X5, Canada
| | - Kin Chan
- Center for Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital and Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5G 1X5, Canada
| | - Mandeep K Gill
- Donnelly Centre and Department of Biochemistry, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Liliana Attisano
- Donnelly Centre and Department of Biochemistry, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Jeffrey L Wrana
- Center for Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital and Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5G 1X5, Canada
| | - Darren A Yuen
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital (Unity Health Toronto) and Department of Medicine, University of Toronto, Toronto, Ontario M5B 1T8, Canada
| |
Collapse
|
23
|
Fernández-Méndez C, Santisteban P. A Critical Balance Between PAX8 and the Hippo Mediator TAZ Determines Sodium/Iodide Symporter Expression and Function. Thyroid 2022; 32:315-325. [PMID: 34726504 DOI: 10.1089/thy.2021.0191] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Background: The Hippo pathway has a fundamental role in tissue homeostasis, but little is known about how this signaling cascade is controlled in the thyroid. PAX8 is an essential driver of thyroid differentiation and is involved in the control of genes crucial for thyroid hormone biosynthesis, including the sodium/iodide symporter (NIS; SLC5A5). A role for the Hippo mediator transcriptional coactivator with PDZ-binding motif (TAZ) as a coactivator of PAX8 to promote thyroglobulin expression has been previously described. Here, we studied the role of TAZ on thyroid differentiation focusing on PAX8-mediated Slc5a5 transcription. Methods: Gene silencing and overexpression assays were performed in rat PCCl3 thyroid follicular cells (TFCs) to determine the role of TAZ in the regulation of Slc5a5. Transcriptional activity of the Hippo mediators was investigated by chromatin immunoprecipitation and promoter-reporter gene activity. Hippo component levels and location were analyzed in PCCl3 cells and in mouse thyroid under different treatment conditions. Results: By suppressing the expression of PAX8 and its binding to the Slc5a5 upstream enhancer, TAZ inhibits Slc5a5 expression, impairing NIS membrane location and activity. Other Hippo effectors such as YAP1 and TEAD1 were not required for the repressor effect of TAZ. We also found an interplay between the Hippo, thyrotropin (TSH), and transforming growth factor β1 (TGFβ) pathways in TFCs. TSH via cyclic adenosine monophosphate activated Hippo signaling pathway and, consequently, TAZ was excluded from the nucleus. We confirmed this in hypothyroid mice, characterized by elevated TSH serum levels, which showed downregulated activation of Hippo signaling in thyroid. Conversely, TAZ nuclear retention was promoted by TGFβ, a potent NIS repressor, and TAZ silencing markedly relieved the TGFβ-induced inhibition of the symporter. Conclusions: We demonstrate that the effects of TAZ are promoter specific, as it functions as a corepressor of PAX8 to modulate Slc5a5 expression in TFCs. Overall, our data place TAZ as an integrator of the different signaling pathways that control NIS expression, pointing to a role for TAZ in thyroid differentiation and identifying the Hippo pathway as a relevant target to recover NIS levels in thyroid cancer cells.
Collapse
Affiliation(s)
- Celia Fernández-Méndez
- Instituto de Investigaciones Biomédicas "Alberto Sols," Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
| | - Pilar Santisteban
- Instituto de Investigaciones Biomédicas "Alberto Sols," Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| |
Collapse
|
24
|
Nalluri SM, Sankhe CS, O'Connor JW, Blanchard PL, Khouri JN, Phan SH, Virgi G, Gomez EW. Crosstalk between ERK and MRTF‐A signaling regulates TGFβ1‐induced epithelial‐mesenchymal transition. J Cell Physiol 2022; 237:2503-2515. [DOI: 10.1002/jcp.30705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 02/08/2022] [Accepted: 02/11/2022] [Indexed: 11/10/2022]
Affiliation(s)
- Sandeep M. Nalluri
- Department of Chemical Engineering The Pennsylvania State University University Park Pennsylvania USA
| | - Chinmay S. Sankhe
- Department of Chemical Engineering The Pennsylvania State University University Park Pennsylvania USA
| | - Joseph W. O'Connor
- Department of Chemical Engineering The Pennsylvania State University University Park Pennsylvania USA
| | - Paul L. Blanchard
- Department of Chemical Engineering The Pennsylvania State University University Park Pennsylvania USA
| | - Joelle N. Khouri
- Department of Chemical Engineering The Pennsylvania State University University Park Pennsylvania USA
| | - Steven H. Phan
- Department of Chemical Engineering The Pennsylvania State University University Park Pennsylvania USA
| | - Gage Virgi
- Department of Chemical Engineering The Pennsylvania State University University Park Pennsylvania USA
| | - Esther W. Gomez
- Department of Chemical Engineering The Pennsylvania State University University Park Pennsylvania USA
- Department of Biomedical Engineering The Pennsylvania State University University Park Pennsylvania USA
| |
Collapse
|
25
|
He X, Tolosa MF, Zhang T, Goru SK, Ulloa Severino L, Misra PS, McEvoy CM, Caldwell L, Szeto SG, Gao F, Chen X, Atin C, Ki V, Vukosa N, Hu C, Zhang J, Yip C, Krizova A, Wrana JL, Yuen DA. Myofibroblast YAP/TAZ activation is a key step in organ fibrogenesis. JCI Insight 2022; 7:146243. [PMID: 35191398 PMCID: PMC8876427 DOI: 10.1172/jci.insight.146243] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 01/12/2022] [Indexed: 12/13/2022] Open
Abstract
Fibrotic diseases account for nearly half of all deaths in the developed world. Despite its importance, the pathogenesis of fibrosis remains poorly understood. Recently, the two mechanosensitive transcription cofactors YAP and TAZ have emerged as important profibrotic regulators in multiple murine tissues. Despite this growing recognition, a number of important questions remain unanswered, including which cell types require YAP/TAZ activation for fibrosis to occur and the time course of this activation. Here, we present a detailed analysis of the role that myofibroblast YAP and TAZ play in organ fibrosis and the kinetics of their activation. Using analyses of cells, as well as multiple murine and human tissues, we demonstrated that myofibroblast YAP and TAZ were activated early after organ injury and that this activation was sustained. We further demonstrated the critical importance of myofibroblast YAP/TAZ in driving progressive scarring in the kidney, lung, and liver, using multiple transgenic models in which YAP and TAZ were either deleted or hyperactivated. Taken together, these data establish the importance of early injury-induced myofibroblast YAP and TAZ activation as a key event driving fibrosis in multiple organs. This information should help guide the development of new antifibrotic YAP/TAZ inhibition strategies.
Collapse
Affiliation(s)
- Xiaolin He
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital (Unity Health Toronto) and Department of Medicine, and
| | - Monica F Tolosa
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital (Unity Health Toronto) and Department of Medicine, and
| | - Tianzhou Zhang
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital (Unity Health Toronto) and Department of Medicine, and
| | - Santosh Kumar Goru
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital (Unity Health Toronto) and Department of Medicine, and
| | - Luisa Ulloa Severino
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital (Unity Health Toronto) and Department of Medicine, and
| | - Paraish S Misra
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital (Unity Health Toronto) and Department of Medicine, and
| | - Caitríona M McEvoy
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital (Unity Health Toronto) and Department of Medicine, and
| | - Lauren Caldwell
- Center for Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital and Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Stephen G Szeto
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital (Unity Health Toronto) and Department of Medicine, and
| | - Feng Gao
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital (Unity Health Toronto) and Department of Medicine, and.,Department of Pathology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China
| | - Xiaolan Chen
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital (Unity Health Toronto) and Department of Medicine, and.,Department of Respiratory and Critical Care Medicine, Beijing Shijitan Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Cassandra Atin
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital (Unity Health Toronto) and Department of Medicine, and
| | - Victoria Ki
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital (Unity Health Toronto) and Department of Medicine, and
| | - Noah Vukosa
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital (Unity Health Toronto) and Department of Medicine, and
| | - Catherine Hu
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital (Unity Health Toronto) and Department of Medicine, and
| | - Johnny Zhang
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital (Unity Health Toronto) and Department of Medicine, and
| | - Christopher Yip
- Faculty of Applied Science and Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Adriana Krizova
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital (Unity Health Toronto) and Department of Medicine, and.,Department of Laboratory Medicine and Pathobiology, St. Michael's Hospital (Unity Health Toronto) and University of Toronto, Toronto, Ontario, Canada
| | - Jeffrey L Wrana
- Center for Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital and Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Darren A Yuen
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital (Unity Health Toronto) and Department of Medicine, and
| |
Collapse
|
26
|
Wang Z, Wang F, Ding XY, Li TE, Wang HY, Gao YH, Wang WJ, Liu YF, Chen XS, Shen KW. Hippo/YAP signaling choreographs the tumor immune microenvironment to promote triple negative breast cancer progression via TAZ/IL-34 axis. Cancer Lett 2021; 527:174-190. [PMID: 34929335 DOI: 10.1016/j.canlet.2021.12.016] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 12/10/2021] [Accepted: 12/13/2021] [Indexed: 12/15/2022]
Abstract
Growing evidence suggests that the bidirectional interactions between cancer cells and their surrounding environment, namely the tumor microenvironment (TME), contribute to cancer progression, metastasis, and resistance to treatment. Intense investigation of the Hippo pathway, which controls multiple central cellular functions in tumorigenesis, was focused on cancer cells. However, the role of the Hippo pathway in modulating tumor-stromal interactions in triple-negative breast cancer remains largely unknown. Therefore, this study focused on revealing the effects of Hippo-YAP/TAZ signaling on the immune microenvironment. Our findings reveal that the activity of the Hippo pathway is associated with worse disease outcomes in TNBC and could increase TAM infiltration through the TAZ/IL-34 axis, leading to an immunosuppressive microenvironment and impairing the treatment efficacy of anti-PD-L1. Thus, the TAZ/IL-34 axis may serve as a novel target for TNBC patients.
Collapse
Affiliation(s)
- Zheng Wang
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Fan Wang
- State Key Laboratory of Oncogenes and Related Genes, Renji-MedX Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xin-Yuan Ding
- Department of Pharmacy, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, 215002, China
| | - Tian-En Li
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, 250012, China
| | - Hao-Yu Wang
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yu-Hao Gao
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Wen-Juan Wang
- Department of Pharmacy, Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Yan-Feng Liu
- State Key Laboratory of Oncogenes and Related Genes, Renji-MedX Stem Cell Research Center, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Xiao-Song Chen
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Kun-Wei Shen
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
27
|
Sharma A, Mir R, Galande S. Epigenetic Regulation of the Wnt/β-Catenin Signaling Pathway in Cancer. Front Genet 2021; 12:681053. [PMID: 34552611 PMCID: PMC8450413 DOI: 10.3389/fgene.2021.681053] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 06/30/2021] [Indexed: 12/12/2022] Open
Abstract
Studies over the past four decades have elucidated the role of Wnt/β-catenin mediated regulation in cell proliferation, differentiation and migration. These processes are fundamental to embryonic development, regeneration potential of tissues, as well as cancer initiation and progression. In this review, we focus on the epigenetic players which influence the Wnt/β-catenin pathway via modulation of its components and coordinated regulation of the Wnt target genes. The role played by crosstalk with other signaling pathways mediating tumorigenesis is also elaborated. The Hippo/YAP pathway is particularly emphasized due to its extensive crosstalk via the Wnt destruction complex. Further, we highlight the recent advances in developing potential therapeutic interventions targeting the epigenetic machinery based on the characterization of these regulatory networks for effective treatment of various cancers and also for regenerative therapies.
Collapse
Affiliation(s)
- Ankita Sharma
- Centre of Excellence in Epigenetics, Department of Biology, Indian Institute of Science Education and Research, Pune, India
| | - Rafeeq Mir
- Centre for Interdisciplinary Research and Innovations, University of Kashmir, Srinagar, India
| | - Sanjeev Galande
- Centre of Excellence in Epigenetics, Department of Biology, Indian Institute of Science Education and Research, Pune, India.,Department of Life Sciences, School of Natural Sciences, Shiv Nadar University, Greater Noida, India
| |
Collapse
|
28
|
Du F, Qi X, Zhang A, Sui F, Wang X, Proud CG, Lin C, Fan X, Li J. MRTF-A-NF-κB/p65 axis-mediated PDL1 transcription and expression contributes to immune evasion of non-small-cell lung cancer via TGF-β. Exp Mol Med 2021; 53:1366-1378. [PMID: 34548615 PMCID: PMC8492728 DOI: 10.1038/s12276-021-00670-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 06/11/2021] [Accepted: 07/05/2021] [Indexed: 11/15/2022] Open
Abstract
PD-L1 is abnormally regulated in many cancers and is critical for immune escape. Fully understanding the regulation of PD-L1 expression is vital for improving the clinical efficacy of relevant anticancer agents. TGF-β plays an important role in the low reactivity of PD-1/PD-L1 antibody immunotherapy. However, it is not very clear whether and how TGF-β affects PD-L1 expression. In the present study, we show that TGF-β upregulates the expression of the transcriptional coactivator MRTF-A in non-small-cell lung cancer cells, which subsequently interacts with NF-κB/p65 rather than SRF to facilitate the binding of NF-κB/p65 to the PDL1 promoter, thereby activating the transcription and expression of PD-L1. This leads to the immune escape of NSCLC cells. This process is dependent on the activation of the TGF-β signaling pathway. In vivo, inhibition of MRTF-A effectively suppresses the growth of lung tumor syngrafts with enrichment of NK and T cells in tumor tissue. Our study defines a new signaling pathway that regulates the transcription and expression of PD-L1 upon TGF-β treatment, which may have a significant impact on research into the application of immunotherapy in treating lung cancer. Better understanding how a critical protein to allow cancer cells to escape immune system may aid in development of improved immunotherapies for lung cancer. The membrane protein PD-L1, expressed on tumor cells, helps them to evade the immune surveillance; existing treatments that block PD-L1 have very low efficacy for some patient partly due to re-expression of PD-L1. Jing Li at Ocean University of China in Qingdao and co-workers found that TGF-β up-regulated in tumor microenvironment boosts PD-L1 transcription and expression in an unusual way, namely, via MRTF-A-NF-κB/p65 axis. Blocking MRTF-A in a mouse model remarkably increased levels of immune cells targeting the tumor and slowed lung tumor growth. These results illuminate the mechanism of immune escape in lung cancers upon TGF-β, which may contribute to develop new treatment to synergize PD-L1 antibody therapy.
Collapse
Affiliation(s)
- Fu Du
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, People's Republic of China
| | - Xin Qi
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, People's Republic of China.,Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, People's Republic of China
| | - Aotong Zhang
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, People's Republic of China
| | - Fanfan Sui
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, People's Republic of China
| | - Xuemin Wang
- South Australian Health & Medical Research Institute, North Terrace, Adelaide, SA, 5000, Australia
| | - Christopher G Proud
- South Australian Health & Medical Research Institute, North Terrace, Adelaide, SA, 5000, Australia.,School of Biological Sciences, University of Adelaide, Adelaide, SA, 5005, Australia
| | - Cunzhi Lin
- Department of Respiratory & Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266555, China
| | - Xinglong Fan
- Department of Thoracic Surgery, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, 266035, China
| | - Jing Li
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, People's Republic of China. .,Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, People's Republic of China. .,Open Studio for Drug Research on Marine Natural Products, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, People's Republic of China.
| |
Collapse
|
29
|
Nucleocytoplasmic Shuttling of the Mechanosensitive Transcription Factors MRTF and YAP /TAZ. Methods Mol Biol 2021; 2299:197-216. [PMID: 34028745 DOI: 10.1007/978-1-0716-1382-5_15] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
Abstract
Myocardin-related transcription factor (MRTF) and the paralogous Hippo pathway effectors Yes-associated protein (YAP) and transcriptional co-activator with PDZ-binding motif (TAZ) are transcriptional co-activators that play pivotal roles in myofibroblast generation and activation, and thus the pathogenesis of organ fibrosis. They are regulated by a variety of chemical and mechanical fibrogenic stimuli, primarily at the level of their nucleocytoplasmic shuttling. In this chapter we describe the tools and protocols that allow for exact, quantitative, and automated determination and analysis of the nucleocytoplasmic distribution of endogenous or heterologously expressed MRTF and YAP/TAZ, measured in large cell populations. Dynamic monitoring of nucleocytoplasmic ratios of transcription factors is a novel and important approach, suitable to address both the structural requirements and the regulatory mechanisms underlying transcription factor traffic and the consequent reprogramming of gene expression during fibrogenesis.
Collapse
|
30
|
Speight P, Rozycki M, Venugopal S, Szászi K, Kofler M, Kapus A. Myocardin-related transcription factor and serum response factor regulate cilium turnover by both transcriptional and local mechanisms. iScience 2021; 24:102739. [PMID: 34278253 PMCID: PMC8261663 DOI: 10.1016/j.isci.2021.102739] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 11/02/2020] [Accepted: 06/15/2021] [Indexed: 12/31/2022] Open
Abstract
Turnover of the primary cilium (PC) is critical for proliferation and tissue homeostasis. Each key component of the PC resorption machinery, the HEF1/Aurora kinase A (AurA)/HDAC6 pathway harbors cis-elements potentially targeted by the transcriptional co-activator myocardin-related transcription factor (MRTF) and/or its partner serum response factor (SRF). Thus we investigated if MRTF and/or SRF regulate PC turnover. Here we show that (1) both MRTF and SRF are indispensable for serum-induced PC resorption, and (2) they act via both transcriptional and local mechanisms. Intriguingly, MRTF and SRF are present in the basal body and/or the PC, and serum facilitates ciliary MRTF recruitment. MRTF promotes the stability and ciliary accumulation of AurA and facilitates SRF phosphorylation. Ciliary SRF interacts with AurA and HDAC6. MRTF also inhibits ciliogenesis. It interacts with and is required for the correct localization of the ciliogenesis modulator CEP290. Thus, MRTF and SRF are critical regulators of PC assembly and/or disassembly, acting both as transcription factors and as PC constituents.
Collapse
Affiliation(s)
- Pam Speight
- Keenan Research Centre for Biomedical Science of the St. Michael's Hospital, University of Toronto, Room 621, 209 Victoria Street, Toronto, ON M5B 1T8, Canada
| | - Matthew Rozycki
- Keenan Research Centre for Biomedical Science of the St. Michael's Hospital, University of Toronto, Room 621, 209 Victoria Street, Toronto, ON M5B 1T8, Canada
| | - Shruthi Venugopal
- Keenan Research Centre for Biomedical Science of the St. Michael's Hospital, University of Toronto, Room 621, 209 Victoria Street, Toronto, ON M5B 1T8, Canada
| | - Katalin Szászi
- Keenan Research Centre for Biomedical Science of the St. Michael's Hospital, University of Toronto, Room 621, 209 Victoria Street, Toronto, ON M5B 1T8, Canada
- Department of Surgery, University of Toronto, Toronto, ON M5B 1T8, Canada
| | - Michael Kofler
- Keenan Research Centre for Biomedical Science of the St. Michael's Hospital, University of Toronto, Room 621, 209 Victoria Street, Toronto, ON M5B 1T8, Canada
| | - András Kapus
- Keenan Research Centre for Biomedical Science of the St. Michael's Hospital, University of Toronto, Room 621, 209 Victoria Street, Toronto, ON M5B 1T8, Canada
- Department of Surgery, University of Toronto, Toronto, ON M5B 1T8, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON M5B 1T8, Canada
| |
Collapse
|
31
|
Li FL, Guan KL. The two sides of Hippo pathway in cancer. Semin Cancer Biol 2021; 85:33-42. [PMID: 34265423 DOI: 10.1016/j.semcancer.2021.07.006] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 07/09/2021] [Accepted: 07/11/2021] [Indexed: 02/08/2023]
Abstract
The Hippo signaling pathway was originally characterized by genetic studies in Drosophila to regulate tissue growth and organ size, and the core components of this pathway are highly conserved in mammals. Studies over the past two decades have revealed critical physiological and pathological functions of the Hippo tumor-suppressor pathway, which is tightly regulated by a broad range of intracellular and extracellular signals. These properties enable the Hippo pathway to serve as an important controller in organismal development and adult tissue homeostasis. Dysregulation of the Hippo signaling has been observed in many cancer types, suggesting the possibility of cancer treatment by targeting the Hippo pathway. The general consensus is that Hippo has tumor suppressor function. However, growing evidence also suggests that the function of the Hippo pathway in malignancy is cancer context dependent as recent studies indicating tumor promoting function of LATS. This article surveys the Hippo pathway signaling mechanisms and then reviews both the tumor suppressing and promoting function of this pathway. A comprehensive understanding of the dual roles of the Hippo pathway in cancer will benefit future therapeutic targeting of the Hippo pathway for cancer treatment.
Collapse
Affiliation(s)
- Fu-Long Li
- Department of Pharmacology, University of California San Diego, La Jolla, CA, USA; Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Kun-Liang Guan
- Department of Pharmacology, University of California San Diego, La Jolla, CA, USA; Moores Cancer Center, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
32
|
Concurrent YAP/TAZ and SMAD signaling mediate vocal fold fibrosis. Sci Rep 2021; 11:13484. [PMID: 34188130 PMCID: PMC8241934 DOI: 10.1038/s41598-021-92871-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 06/10/2021] [Indexed: 01/17/2023] Open
Abstract
Vocal fold (VF) fibrosis is a major cause of intractable voice-related disability and reduced quality of life. Excision of fibrotic regions is suboptimal and associated with scar recurrence and/or further iatrogenic damage. Non-surgical interventions are limited, putatively related to limited insight regarding biochemical events underlying fibrosis, and downstream, the lack of therapeutic targets. YAP/TAZ integrates diverse cell signaling events and interacts with signaling pathways related to fibrosis, including the TGF-β/SMAD pathway. We investigated the expression of YAP/TAZ following vocal fold injury in vivo as well as the effects of TGF-β1 on YAP/TAZ activity in human vocal fold fibroblasts, fibroblast-myofibroblast transition, and TGF-β/SMAD signaling. Iatrogenic injury increased nuclear localization of YAP and TAZ in fibrotic rat vocal folds. In vitro, TGF-β1 activated YAP and TAZ in human VF fibroblasts, and inhibition of YAP/TAZ reversed TGF-β1-stimulated fibroplastic gene upregulation. Additionally, TGF-β1 induced localization of YAP and TAZ in close proximity to SMAD2/3, and nuclear accumulation of SMAD2/3 was inhibited by a YAP/TAZ inhibitor. Collectively, YAP and TAZ were synergistically activated with the TGF-β/SMAD pathway, and likely essential for the fibroplastic phenotypic shift in VF fibroblasts. Based on these data, YAP/TAZ may evolve as an attractive therapeutic target for VF fibrosis.
Collapse
|
33
|
Miranda MZ, Lichner Z, Szászi K, Kapus A. MRTF: Basic Biology and Role in Kidney Disease. Int J Mol Sci 2021; 22:ijms22116040. [PMID: 34204945 PMCID: PMC8199744 DOI: 10.3390/ijms22116040] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 05/21/2021] [Accepted: 05/30/2021] [Indexed: 12/23/2022] Open
Abstract
A lesser known but crucially important downstream effect of Rho family GTPases is the regulation of gene expression. This major role is mediated via the cytoskeleton, the organization of which dictates the nucleocytoplasmic shuttling of a set of transcription factors. Central among these is myocardin-related transcription factor (MRTF), which upon actin polymerization translocates to the nucleus and binds to its cognate partner, serum response factor (SRF). The MRTF/SRF complex then drives a large cohort of genes involved in cytoskeleton remodeling, contractility, extracellular matrix organization and many other processes. Accordingly, MRTF, activated by a variety of mechanical and chemical stimuli, affects a plethora of functions with physiological and pathological relevance. These include cell motility, development, metabolism and thus metastasis formation, inflammatory responses and—predominantly-organ fibrosis. The aim of this review is twofold: to provide an up-to-date summary about the basic biology and regulation of this versatile transcriptional coactivator; and to highlight its principal involvement in the pathobiology of kidney disease. Acting through both direct transcriptional and epigenetic mechanisms, MRTF plays a key (yet not fully appreciated) role in the induction of a profibrotic epithelial phenotype (PEP) as well as in fibroblast-myofibroblast transition, prime pathomechanisms in chronic kidney disease and renal fibrosis.
Collapse
Affiliation(s)
- Maria Zena Miranda
- Keenan Research Centre for Biomedical Science of the St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada; (M.Z.M.); (Z.L.); (K.S.)
| | - Zsuzsanna Lichner
- Keenan Research Centre for Biomedical Science of the St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada; (M.Z.M.); (Z.L.); (K.S.)
| | - Katalin Szászi
- Keenan Research Centre for Biomedical Science of the St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada; (M.Z.M.); (Z.L.); (K.S.)
- Department of Surgery, University of Toronto, Toronto, ON M5T 1P5, Canada
| | - András Kapus
- Keenan Research Centre for Biomedical Science of the St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada; (M.Z.M.); (Z.L.); (K.S.)
- Department of Surgery, University of Toronto, Toronto, ON M5T 1P5, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
- Correspondence:
| |
Collapse
|
34
|
SKI activates the Hippo pathway via LIMD1 to inhibit cardiac fibroblast activation. Basic Res Cardiol 2021; 116:25. [PMID: 33847835 PMCID: PMC8043893 DOI: 10.1007/s00395-021-00865-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 03/24/2021] [Indexed: 01/14/2023]
Abstract
We have previously shown that overexpression of SKI, an endogenous TGF-β1 repressor, deactivates the pro-fibrotic myofibroblast phenotype in the heart. We now show that SKI also functions independently of SMAD/TGF-β signaling, by activating the Hippo tumor-suppressor pathway and inhibiting the Transcriptional co-Activator with PDZ-binding motif (TAZ or WWTR1). The mechanism(s) by which SKI targets TAZ to inhibit cardiac fibroblast activation and fibrogenesis remain undefined. A rat model of post-myocardial infarction was used to examine the expression of TAZ during acute fibrogenesis and chronic heart failure. Results were then corroborated with primary rat cardiac fibroblast cell culture performed both on plastic and on inert elastic substrates, along with the use of siRNA and adenoviral expression vectors for active forms of SKI, YAP, and TAZ. Gene expression was examined by qPCR and luciferase assays, while protein expression was examined by immunoblotting and fluorescence microscopy. Cell phenotype was further assessed by functional assays. Finally, to elucidate SKI’s effects on Hippo signaling, the SKI and TAZ interactomes were captured in human cardiac fibroblasts using BioID2 and mass spectrometry. Potential interactors were investigated in vitro to reveal novel mechanisms of action for SKI. In vitro assays on elastic substrates revealed the ability of TAZ to overcome environmental stimuli and induce the activation of hypersynthetic cardiac myofibroblasts. Further cell-based assays demonstrated that SKI causes specific proteasomal degradation of TAZ, but not YAP, and shifts actin cytoskeleton dynamics to inhibit myofibroblast activation. These findings were supported by identifying the bi-phasic expression of TAZ in vivo during post-MI remodeling and fibrosis. BioID2-based interactomics in human cardiac fibroblasts suggest that SKI interacts with actin-modifying proteins and with LIM Domain-containing protein 1 (LIMD1), a negative regulator of Hippo signaling. Furthermore, we found that LATS2 interacts with TAZ, whereas LATS1 does not, and that LATS2 knockdown prevented TAZ downregulation with SKI overexpression. Our findings indicate that SKI’s capacity to regulate cardiac fibroblast activation is mediated, in part, by Hippo signaling. We postulate that the interaction between SKI and TAZ in cardiac fibroblasts is arbitrated by LIMD1, an important intermediary in focal adhesion-associated signaling pathways. This study contributes to the understanding of the unique physiology of cardiac fibroblasts, and of the relationship between SKI expression and cell phenotype.
Collapse
|
35
|
Sprenkeler EGG, Guenther C, Faisal I, Kuijpers TW, Fagerholm SC. Molecular Mechanisms of Leukocyte Migration and Its Potential Targeting-Lessons Learned From MKL1/SRF-Related Primary Immunodeficiency Diseases. Front Immunol 2021; 12:615477. [PMID: 33692789 PMCID: PMC7938309 DOI: 10.3389/fimmu.2021.615477] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 01/04/2021] [Indexed: 01/22/2023] Open
Abstract
Megakaryoblastic leukemia 1 (MKL1) deficiency is one of the most recently discovered primary immunodeficiencies (PIDs) caused by cytoskeletal abnormalities. These immunological “actinopathies” primarily affect hematopoietic cells, resulting in defects in both the innate immune system (phagocyte defects) and adaptive immune system (T-cell and B-cell defects). MKL1 is a transcriptional coactivator that operates together with serum response factor (SRF) to regulate gene transcription. The MKL/SRF pathway has been originally described to have important functions in actin regulation in cells. Recent results indicate that MKL1 also has very important roles in immune cells, and that MKL1 deficiency results in an immunodeficiency affecting the migration and function of primarily myeloid cells such as neutrophils. Interestingly, several actinopathies are caused by mutations in genes which are recognized MKL(1/2)-dependent SRF-target genes, namely ACTB, WIPF1, WDR1, and MSN. Here we summarize these and related (ARPC1B) actinopathies and their effects on immune cell function, especially focusing on their effects on leukocyte adhesion and migration. Furthermore, we summarize recent therapeutic efforts targeting the MKL/SRF pathway in disease.
Collapse
Affiliation(s)
- Evelien G G Sprenkeler
- Department of Blood Cell Research, Sanquin Research, Amsterdam University Medical Center (AUMC), University of Amsterdam, Amsterdam, Netherlands.,Department of Pediatric Immunology, Rheumatology, and Infectious Diseases, Emma Children's Hospital, Amsterdam University Medical Center (AUMC), University of Amsterdam, Amsterdam, Netherlands
| | - Carla Guenther
- Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Imrul Faisal
- Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Taco W Kuijpers
- Department of Blood Cell Research, Sanquin Research, Amsterdam University Medical Center (AUMC), University of Amsterdam, Amsterdam, Netherlands.,Department of Pediatric Immunology, Rheumatology, and Infectious Diseases, Emma Children's Hospital, Amsterdam University Medical Center (AUMC), University of Amsterdam, Amsterdam, Netherlands
| | - Susanna C Fagerholm
- Molecular and Integrative Biosciences Research Programme, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| |
Collapse
|
36
|
Wang D, Rabhi N, Yet SF, Farmer SR, Layne MD. Aortic carboxypeptidase-like protein regulates vascular adventitial progenitor and fibroblast differentiation through myocardin related transcription factor A. Sci Rep 2021; 11:3948. [PMID: 33597582 PMCID: PMC7889889 DOI: 10.1038/s41598-021-82941-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 01/25/2021] [Indexed: 02/06/2023] Open
Abstract
The vascular adventitia contains numerous cell types including fibroblasts, adipocytes, inflammatory cells, and progenitors embedded within a complex extracellular matrix (ECM) network. In response to vascular injury, adventitial progenitors and fibroblasts become activated and exhibit increased proliferative capacity and differentiate into contractile cells that remodel the ECM. These processes can lead to vascular fibrosis and disease progression. Our previous work established that the ECM protein aortic carboxypeptidase-like protein (ACLP) promotes fibrotic remodeling in the lung and is activated by vascular injury. It is currently unknown what controls vascular adventitial cell differentiation and if ACLP has a role in this process. Using purified mouse aortic adventitia Sca1+ progenitors, ACLP repressed stem cell markers (CD34, KLF4) and upregulated smooth muscle actin (SMA) and collagen I expression. ACLP enhanced myocardin-related transcription factor A (MRTFA) activity in adventitial cells by promoting MRTFA nuclear translocation. Sca1 cells from MRTFA-null mice exhibited reduced SMA and collagen expression induced by ACLP, indicating Sca1 cell differentiation is regulated in part by the ACLP-MRTFA axis. We determined that ACLP induced vessel contraction and increased adventitial collagen in an explant model. Collectively these studies identified ACLP as a mediator of adventitial cellular differentiation, which may result in pathological vessel remodeling.
Collapse
Affiliation(s)
- Dahai Wang
- Department of Biochemistry, Boston University School of Medicine, 72 E. Concord St, Boston, MA, 02118, USA.,Department of Hematology, Boston Children's Hospital, Boston, MA, USA
| | - Nabil Rabhi
- Department of Biochemistry, Boston University School of Medicine, 72 E. Concord St, Boston, MA, 02118, USA
| | - Shaw-Fang Yet
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, 35053, Taiwan
| | - Stephen R Farmer
- Department of Biochemistry, Boston University School of Medicine, 72 E. Concord St, Boston, MA, 02118, USA
| | - Matthew D Layne
- Department of Biochemistry, Boston University School of Medicine, 72 E. Concord St, Boston, MA, 02118, USA.
| |
Collapse
|
37
|
LeBlanc L, Ramirez N, Kim J. Context-dependent roles of YAP/TAZ in stem cell fates and cancer. Cell Mol Life Sci 2021; 78:4201-4219. [PMID: 33582842 PMCID: PMC8164607 DOI: 10.1007/s00018-021-03781-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 12/30/2020] [Accepted: 01/28/2021] [Indexed: 02/06/2023]
Abstract
Hippo effectors YAP and TAZ control cell fate and survival through various mechanisms, including transcriptional regulation of key genes. However, much of this research has been marked by conflicting results, as well as controversy over whether YAP and TAZ are redundant. A substantial portion of the discordance stems from their contradictory roles in stem cell self-renewal vs. differentiation and cancer cell survival vs. apoptosis. In this review, we present an overview of the multiple context-dependent functions of YAP and TAZ in regulating cell fate decisions in stem cells and organoids, as well as their mechanisms of controlling programmed cell death pathways in cancer.
Collapse
Affiliation(s)
- Lucy LeBlanc
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, 78712, USA. .,Interdisciplinary Life Sciences Graduate Program, The University of Texas at Austin, Austin, TX, 78712, USA.
| | - Nereida Ramirez
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, 78712, USA.,Harvard Medical School, 25 Shattuck St, Boston, MA, 02115, USA
| | - Jonghwan Kim
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, 78712, USA. .,Interdisciplinary Life Sciences Graduate Program, The University of Texas at Austin, Austin, TX, 78712, USA. .,Center for Systems and Synthetic Biology, The University of Texas at Austin, Austin, TX, 78712, USA.
| |
Collapse
|
38
|
YAP/TAZ inhibition reduces metastatic potential of Ewing sarcoma cells. Oncogenesis 2021; 10:2. [PMID: 33419969 PMCID: PMC7794350 DOI: 10.1038/s41389-020-00294-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 12/04/2020] [Accepted: 12/08/2020] [Indexed: 12/15/2022] Open
Abstract
Ewing sarcoma (EwS) is a highly metastatic bone cancer characterized by the ETS fusion oncoprotein EWS-FLI1. EwS cells are phenotypically highly plastic and switch between functionally distinct cell states dependent on EWS-FLI1 fluctuations. Whereas EWS-FLI1high cells proliferate, EWS-FLI1low cells are migratory and invasive. Recently, we reported activation of MRTFB and TEAD, effectors of RhoA and Hippo signalling, upon low EWS-FLI1, orchestrating key steps of the EwS migratory gene expression program. TEAD and its co-activators YAP and TAZ are commonly overexpressed in cancer, providing attractive therapeutic targets. We find TAZ levels to increase in the migratory EWS-FLI1low state and to associate with adverse prognosis in EwS patients. We tested the effects of the potent YAP/TAZ/TEAD complex inhibitor verteporfin on EwS cell migration in vitro and on metastasis in vivo. Verteporfin suppressed expression of EWS-FLI1 regulated cytoskeletal genes involved in actin signalling to the extracellular matrix, effectively blocked F-actin and focal-adhesion assembly and inhibited EwS cell migration at submicromolar concentrations. In a mouse EwS xenograft model, verteporfin treatment reduced relapses at the surgical site and delayed lung metastasis. These data suggest that YAP/TAZ pathway inhibition may prevent EwS cell dissemination and metastasis, justifying further preclinical development of YAP/TAZ inhibitors for EwS treatment.
Collapse
|
39
|
Heng BC, Zhang X, Aubel D, Bai Y, Li X, Wei Y, Fussenegger M, Deng X. An overview of signaling pathways regulating YAP/TAZ activity. Cell Mol Life Sci 2021; 78:497-512. [PMID: 32748155 PMCID: PMC11071991 DOI: 10.1007/s00018-020-03579-8] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 03/07/2020] [Accepted: 06/22/2020] [Indexed: 12/11/2022]
Abstract
YAP and TAZ are ubiquitously expressed homologous proteins originally identified as penultimate effectors of the Hippo signaling pathway, which plays a key role in maintaining mammalian tissue/organ size. Presently, it is known that YAP/TAZ also interact with various non-Hippo signaling pathways, and have diverse roles in multiple biological processes, including cell proliferation, tissue regeneration, cell lineage fate determination, tumorigenesis, and mechanosensing. In this review, we first examine the various microenvironmental cues and signaling pathways that regulate YAP/TAZ activation, through the Hippo and non-Hippo signaling pathways. This is followed by a brief summary of the interactions of YAP/TAZ with TEAD1-4 and a diverse array of other non-TEAD transcription factors. Finally, we offer a critical perspective on how increasing knowledge of the regulatory mechanisms of YAP/TAZ signaling might open the door to novel therapeutic applications in the interrelated fields of biomaterials, tissue engineering, regenerative medicine and synthetic biology.
Collapse
Affiliation(s)
- Boon Chin Heng
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, 100081, People's Republic of China
- Faculty of Science and Technology, Sunway University, Selangor Darul Ehsan, Malaysia
| | - Xuehui Zhang
- Department of Dental Materials & Dental Medical Devices Testing Center, Peking University School and Hospital of Stomatology, Beijing, 100081, People's Republic of China
- National Engineering Laboratory for Digital and Material Technology of Stomatology, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials, Peking University School and Hospital of Stomatology, Beijing, 100081, People's Republic of China
| | - Dominique Aubel
- IUTA, Departement Genie Biologique, Universite, Claude Bernard Lyon 1, Villeurbanne Cedex, France
| | - Yunyang Bai
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, People's Republic of China
| | - Xiaochan Li
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, People's Republic of China
| | - Yan Wei
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, People's Republic of China
| | - Martin Fussenegger
- Department of Biosystems Science and Engineering, ETH-Zurich, Mattenstrasse 26, Basel, 4058, Switzerland.
| | - Xuliang Deng
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, People's Republic of China.
- National Engineering Laboratory for Digital and Material Technology of Stomatology, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials, Peking University School and Hospital of Stomatology, Beijing, 100081, People's Republic of China.
| |
Collapse
|
40
|
Recent Advances in Understandings Towards Pathogenesis and Treatment for Intrauterine Adhesion and Disruptive Insights from Single-Cell Analysis. Reprod Sci 2020; 28:1812-1826. [PMID: 33125685 PMCID: PMC8189970 DOI: 10.1007/s43032-020-00343-y] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 10/01/2020] [Indexed: 12/22/2022]
Abstract
Intrauterine adhesion is a major cause of menstrual irregularities, infertility, and recurrent pregnancy losses and the progress towards its amelioration and therapy is slow and unsatisfactory. We aim to summarize and evaluate the current treatment progress and research methods for intrauterine adhesion. We conducted literature review in January 2020 by searching articles at PubMed on prevention and treatment, pathogenesis, the repair of other tissues/organs, cell plasticity, and the stem cell–related therapies for intrauterine adhesion. A total of 110 articles were selected for review. Uterine cell heterogeneity, expression profile, and cell-cell interaction were investigated based on scRNA-seq of uterus provided by Human Cell Landscape (HCL) project. Previous knowledge on intrauterine adhesion (IUA) pathogenesis was mostly derived from correlation studies by differentially expressed genes between endometrial tissue of intrauterine adhesion patients/animal models and normal endometrial tissue. Although the TGF-β1/SMAD pathway was suggested as the key driver for IUA pathogenesis, uterine cell heterogeneity and distinct expression profile among different cell types highlighted the importance of single-cell investigations. Cell-cell interaction in the uterus revealed the central hub of endothelial cells interacting with other cells, with endothelial cells in endothelial to mesenchymal transition and fibroblasts as the strongest interaction partners. The potential of stem cell–related therapies appeared promising, yet suffers from largely animal studies and nonstandard study design. The need to dissect the roles of endometrial cells, endothelial cells, and fibroblasts and their interaction is evident in order to elucidate the molecular and cellular mechanisms in both intrauterine adhesion pathogenesis and treatment.
Collapse
|
41
|
Freeberg MAT, Perelas A, Rebman JK, Phipps RP, Thatcher TH, Sime PJ. Mechanical Feed-Forward Loops Contribute to Idiopathic Pulmonary Fibrosis. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 191:18-25. [PMID: 33031756 PMCID: PMC7768346 DOI: 10.1016/j.ajpath.2020.09.008] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 08/25/2020] [Accepted: 09/08/2020] [Indexed: 12/11/2022]
Abstract
Idiopathic pulmonary fibrosis is a progressive scarring disease characterized by extracellular matrix accumulation and altered mechanical properties of lung tissue. Recent studies support the hypothesis that these compositional and mechanical changes create a progressive feed-forward loop in which enhanced matrix deposition and tissue stiffening contribute to fibroblast and myofibroblast differentiation and activation, which further perpetuates matrix production and stiffening. The biomechanical properties of tissues are sensed and responded to by mechanotransduction pathways that facilitate sensing of changes in mechanical cues by tissue resident cells and convert the mechanical signals into downstream biochemical signals. Although our understanding of mechanotransduction pathways associated with pulmonary fibrosis remains incomplete, recent progress has allowed us to begin to elucidate the specific mechanisms supporting fibrotic feed-forward loops. The mechanosensors discussed here include integrins, Piezo channels, transient receptor potential channels, and nonselective ion channels. Also discussed are downstream transcription factors, including myocardin-related transcription factor and Yes-associated protein/transcriptional coactivator with PDZ-binding motif. This review describes mechanosensors and mechanotransduction pathways associated with fibrosis progression and highlights promising therapeutic insights.
Collapse
Affiliation(s)
- Margaret A T Freeberg
- Division of Pulmonary Disease and Critical Care Medicine, Virginia Commonwealth University, Richmond, Virginia
| | - Apostolos Perelas
- Division of Pulmonary Disease and Critical Care Medicine, Virginia Commonwealth University, Richmond, Virginia
| | - Jane K Rebman
- Division of Pulmonary Disease and Critical Care Medicine, Virginia Commonwealth University, Richmond, Virginia
| | | | - Thomas H Thatcher
- Division of Pulmonary Disease and Critical Care Medicine, Virginia Commonwealth University, Richmond, Virginia
| | - Patricia J Sime
- Division of Pulmonary Disease and Critical Care Medicine, Virginia Commonwealth University, Richmond, Virginia.
| |
Collapse
|
42
|
Reggiani F, Gobbi G, Ciarrocchi A, Sancisi V. YAP and TAZ Are Not Identical Twins. Trends Biochem Sci 2020; 46:154-168. [PMID: 32981815 DOI: 10.1016/j.tibs.2020.08.012] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 08/13/2020] [Accepted: 08/27/2020] [Indexed: 12/12/2022]
Abstract
Yes-associated protein (YAP) and TAZ (WW domain containing transcription regulator 1, or WWTR1) are paralog transcriptional regulators, able to integrate mechanical, metabolic, and signaling inputs to regulate cell growth and differentiation during development and neoplastic progression. YAP and TAZ hold common and distinctive structural features, reflecting only partially overlapping regulatory mechanisms. The two paralogs interact with both shared and specific transcriptional partners and control nonidentical transcriptional programs. Although most of the available literature considers YAP and TAZ as functionally redundant, they play distinctive or even contrasting roles in different contexts. The issue of their divergent roles is currently underexplored but holds fundamental implications for mechanistic and translational studies. Here, we aim to review the available literature on the biological functions of YAP and TAZ, highlighting differential roles that distinguish these two paralogues.
Collapse
Affiliation(s)
- Francesca Reggiani
- Laboratory of Translational Research, Azienda USL- IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Giulia Gobbi
- Laboratory of Translational Research, Azienda USL- IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Alessia Ciarrocchi
- Laboratory of Translational Research, Azienda USL- IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Valentina Sancisi
- Laboratory of Translational Research, Azienda USL- IRCCS di Reggio Emilia, Reggio Emilia, Italy.
| |
Collapse
|
43
|
Hiepen C, Mendez PL, Knaus P. It Takes Two to Tango: Endothelial TGFβ/BMP Signaling Crosstalk with Mechanobiology. Cells 2020; 9:E1965. [PMID: 32858894 PMCID: PMC7564048 DOI: 10.3390/cells9091965] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 08/19/2020] [Accepted: 08/22/2020] [Indexed: 02/06/2023] Open
Abstract
Bone morphogenetic proteins (BMPs) are members of the transforming growth factor-beta (TGFβ) superfamily of cytokines. While some ligand members are potent inducers of angiogenesis, others promote vascular homeostasis. However, the precise understanding of the molecular mechanisms underlying these functions is still a growing research field. In bone, the tissue in which BMPs were first discovered, crosstalk of TGFβ/BMP signaling with mechanobiology is well understood. Likewise, the endothelium represents a tissue that is constantly exposed to multiple mechanical triggers, such as wall shear stress, elicited by blood flow or strain, and tension from the surrounding cells and to the extracellular matrix. To integrate mechanical stimuli, the cytoskeleton plays a pivotal role in the transduction of these forces in endothelial cells. Importantly, mechanical forces integrate on several levels of the TGFβ/BMP pathway, such as receptors and SMADs, but also global cell-architecture and nuclear chromatin re-organization. Here, we summarize the current literature on crosstalk mechanisms between biochemical cues elicited by TGFβ/BMP growth factors and mechanical cues, as shear stress or matrix stiffness that collectively orchestrate endothelial function. We focus on the different subcellular compartments in which the forces are sensed and integrated into the TGFβ/BMP growth factor signaling.
Collapse
Affiliation(s)
| | | | - Petra Knaus
- Knaus-Lab/Signal Transduction, Institute for Chemistry and Biochemistry, Freie Universitaet Berlin, 14195 Berlin, Germany; (C.H.); (P.-L.M.)
| |
Collapse
|
44
|
Kovar H, Bierbaumer L, Radic-Sarikas B. The YAP/TAZ Pathway in Osteogenesis and Bone Sarcoma Pathogenesis. Cells 2020; 9:E972. [PMID: 32326412 PMCID: PMC7227004 DOI: 10.3390/cells9040972] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 04/10/2020] [Accepted: 04/11/2020] [Indexed: 12/14/2022] Open
Abstract
YAP and TAZ are intracellular messengers communicating multiple interacting extracellular biophysical and biochemical cues to the transcription apparatus in the nucleus and back to the cell/tissue microenvironment interface through the regulation of cytoskeletal and extracellular matrix components. Their activity is negatively and positively controlled by multiple phosphorylation events. Phenotypically, they serve an important role in cellular plasticity and lineage determination during development. As they regulate self-renewal, proliferation, migration, invasion and differentiation of stem cells, perturbed expression of YAP/TAZ signaling components play important roles in tumorigenesis and metastasis. Despite their high structural similarity, YAP and TAZ are functionally not identical and may play distinct cell type and differentiation stage-specific roles mediated by a diversity of downstream effectors and upstream regulatory molecules. However, YAP and TAZ are frequently looked at as functionally redundant and are not sufficiently discriminated in the scientific literature. As the extracellular matrix composition and mechanosignaling are of particular relevance in bone formation during embryogenesis, post-natal bone elongation and bone regeneration, YAP/TAZ are believed to have critical functions in these processes. Depending on the differentiation stage of mesenchymal stem cells during endochondral bone development, YAP and TAZ serve distinct roles, which are also reflected in bone tumors arising from the mesenchymal lineage at different developmental stages. Efforts to clinically translate the wealth of available knowledge of the pathway for cancer diagnostic and therapeutic purposes focus mainly on YAP and TAZ expression and their role as transcriptional co-activators of TEAD transcription factors but rarely consider the expression and activity of pathway modulatory components and other transcriptional partners of YAP and TAZ. As there is a growing body of evidence for YAP and TAZ as potential therapeutic targets in several cancers, we here interrogate the applicability of this concept to bone tumors. To this end, this review aims to summarize our current knowledge of YAP and TAZ in cell plasticity, normal bone development and bone cancer.
Collapse
Affiliation(s)
- Heinrich Kovar
- St. Anna Children’s Cancer Research Institute, 1090 Vienna, Austria; (L.B.); (B.R.-S.)
- Department of Pediatrics, Medical University Vienna, 1090 Vienna, Austria
| | - Lisa Bierbaumer
- St. Anna Children’s Cancer Research Institute, 1090 Vienna, Austria; (L.B.); (B.R.-S.)
| | - Branka Radic-Sarikas
- St. Anna Children’s Cancer Research Institute, 1090 Vienna, Austria; (L.B.); (B.R.-S.)
| |
Collapse
|
45
|
Abstract
PURPOSE OF REVIEW Preclinical data suggests that transforming growth factor-β (TGF-β) is arguably the most potent profibrotic growth factor in kidney injury. Despite this, recent clinical trials targeting TGF-β have been disappointing. These negative studies suggest that TGF-β signaling in the injured kidney might be more complicated than originally thought. This review examines recent studies that expand our understanding of how this pleiotropic growth factor affects renal injury. RECENT FINDINGS There are recent studies showing new mechanisms whereby TGF-β can mediate injury (e.g. epigenetic effects, macrophage chemoattractant). However, more significant are the increasing reports on cross-talk between TGF-β signaling and other pathways relevant to renal injury such as Wnt/β-catenin, YAP/TAZ (transcriptional coactivator with PDZ-binding motif), and klotho/FGF23. TGF-β clearly alters the response to injury, not just by direct transcriptional changes on target cells, but also through effects on other signaling pathways. In T cells and tubular epithelial cells, some of these TGF-β-mediated changes are potentially beneficial. SUMMARY It is unlikely that inhibition of TGF-β per se will be a successful antifibrotic strategy, but a better understanding of TGF-β's actions may reveal promising downstream targets or modulators of signaling to target therapeutically for chronic kidney disease.
Collapse
|
46
|
Liu W, Lu X, Shi P, Yang G, Zhou Z, Li W, Mao X, Jiang D, Chen C. TNF-α increases breast cancer stem-like cells through up-regulating TAZ expression via the non-canonical NF-κB pathway. Sci Rep 2020; 10:1804. [PMID: 32019974 PMCID: PMC7000832 DOI: 10.1038/s41598-020-58642-y] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 01/15/2020] [Indexed: 12/18/2022] Open
Abstract
Breast cancer patients often suffer from disease relapse and metastasis due to the presence of breast cancer stem-like cells (BCSCs). Numerous studies have reported that high levels of inflammatory factors, including tumor necrosis factor alpha (TNF-α), promote BCSCs. However, the mechanism by which TNF-α promotes BCSCs is unclear. In this study, we demonstrate that TNF-α up-regulates TAZ, a transcriptional co-activator promoting BCSC self-renewal capacity in human breast cancer cell lines. Depletion of TAZ abrogated the increase in BCSCs mediated by TNF-α. TAZ is induced by TNF-α through the non-canonical NF-κB pathway, and our findings suggest that TAZ plays a crucial role in inflammatory factor-promoted breast cancer stemness and could serve as a promising therapeutic target.
Collapse
Affiliation(s)
- Wenjing Liu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China
- University of the Chinese Academy of Sciences, Beijing, 101407, China
- Medical Faculty of Kunming University of Science and Technology, Kunming, Yunnan, 650500, China
| | - Xiaoqing Lu
- Department of breast surgery, The second hospital of Shanxi medical University, Taiyuan, 030071, China
| | - Peiguo Shi
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China
| | - Guangxi Yang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China
| | - Zhongmei Zhou
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China
| | - Wei Li
- Medical Faculty of Kunming University of Science and Technology, Kunming, Yunnan, 650500, China
- Department of Urology of the First People's Hospital of Yunnan Province, Kunming, 650032, China
| | - Xiaoyun Mao
- Breast surgery, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China.
| | - Dewei Jiang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China.
| | - Ceshi Chen
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China.
- KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China.
| |
Collapse
|
47
|
Mangum KD, Freeman EJ, Magin JC, Taylor JM, Mack CP. Transcriptional and posttranscriptional regulation of the SMC-selective blood pressure-associated gene, ARHGAP42. Am J Physiol Heart Circ Physiol 2020; 318:H413-H424. [PMID: 31886719 PMCID: PMC7052622 DOI: 10.1152/ajpheart.00143.2019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 12/23/2019] [Accepted: 12/26/2019] [Indexed: 12/19/2022]
Abstract
We previously showed that ARHGAP42 is a smooth muscle cell (SMC)-selective, RhoA-specific GTPase activating protein that regulates blood pressure and that a minor allele single nucleotide variation within a DNAse hypersensitive regulatory element in intron1 (Int1DHS) increased ARHGAP42 expression by promoting serum response factor binding. The goal of the current study was to identify additional transcriptional and posttranscriptional mechanisms that control ARHGAP42 expression. Using deletion/mutation, gel shift, and chromatin immunoprecipitation experiments, we showed that recombination signal binding protein for immunoglobulin κ-J region (RBPJ) and TEA domain family member 1 (TEAD1) binding to a conserved core region was required for full IntDHS transcriptional activity. Importantly, overexpression of the notch intracellular domain (NICD) or plating SMCs on recombinant jagged-1 increased IntDHS activity and endogenous ARHGAP42 expression while siRNA-mediated knockdown of TEAD1 inhibited ARHGAP42 mRNA levels. Re-chromatin immunoprecipitation experiments indicated that RBPJ and TEAD1 were bound to the Int1DHS enhancer at the same time, and coimmunoprecipitation assays indicated that these factors interacted physically. Our results also suggest TEAD1 and RBPJ bound cooperatively to the Int1DHS and that the presence of TEAD1 promoted the recruitment of NICD by RBPJ. Finally, we showed that ARHGAP42 expression was inhibited by micro-RNA 505 (miR505) which interacted with the ARHGAP42 3'-untranslated region (UTR) to facilitate its degradation and by AK124326, a long noncoding RNA that overlaps with the ARHGAP42 transcription start site on the opposite DNA strand. Since siRNA-mediated depletion of AK124326 was associated with increased H3K9 acetylation and RNA Pol-II binding at the ARHGAP42 gene, it is likely that AK124326 inhibits ARHGAP42 transcription.NEW & NOTEWORTHY First, RBPJ and TEAD1 converge at an intronic enhancer to regulate ARHGAP42 expression in SMCs. Second, TEAD1 and RBPJ interact physically and bind cooperatively to the ARHGAP42 enhancer. Third, miR505 interacts with the ARHGAP42 3'-UTR to facilitate its degradation. Finally, LncRNA, AK124326, inhibits ARHGAP42 transcription.
Collapse
Affiliation(s)
- Kevin D Mangum
- Department of Pathology and the McAllister Heart Institute, University of North Carolina at Chapel Hill
| | - Emily J Freeman
- Department of Pathology and the McAllister Heart Institute, University of North Carolina at Chapel Hill
| | - Justin C Magin
- Department of Pathology and the McAllister Heart Institute, University of North Carolina at Chapel Hill
| | - Joan M Taylor
- Department of Pathology and the McAllister Heart Institute, University of North Carolina at Chapel Hill
| | - Christopher P Mack
- Department of Pathology and the McAllister Heart Institute, University of North Carolina at Chapel Hill
| |
Collapse
|
48
|
Haak AJ, Ducharme MT, Diaz Espinosa AM, Tschumperlin DJ. Targeting GPCR Signaling for Idiopathic Pulmonary Fibrosis Therapies. Trends Pharmacol Sci 2020; 41:172-182. [PMID: 32008852 DOI: 10.1016/j.tips.2019.12.008] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 12/10/2019] [Accepted: 12/30/2019] [Indexed: 12/11/2022]
Abstract
A variety of G protein-coupled receptors (GPCRs) have been implicated in the pathogenesis of pulmonary fibrosis, largely through their promotion of profibrotic fibroblast activation. By contrast, recent work has highlighted the beneficial effects of Gαs-coupled GPCRs on reducing fibroblast activation and fibrosis. This review highlights how fibrosis-promoting and -inhibiting GPCR signaling converges on downstream signaling and transcriptional effectors, and how the diversity and dynamics of GPCR expression challenge efforts to identify effective therapies for idiopathic pulmonary fibrosis (IPF). Next-generation strategies to overcome these challenges, focusing on target selection, polypharmacology, and personalized medicine approaches, are discussed as a path towards more effective GPCR-targeted therapies for pulmonary fibrosis.
Collapse
Affiliation(s)
- Andrew J Haak
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA.
| | - Merrick T Ducharme
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Ana M Diaz Espinosa
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Daniel J Tschumperlin
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
49
|
Kegelman CD, Coulombe JC, Jordan KM, Horan DJ, Qin L, Robling AG, Ferguson VL, Bellido TM, Boerckel JD. YAP and TAZ Mediate Osteocyte Perilacunar/Canalicular Remodeling. J Bone Miner Res 2020; 35:196-210. [PMID: 31610061 PMCID: PMC7066596 DOI: 10.1002/jbmr.3876] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 08/13/2019] [Accepted: 09/07/2019] [Indexed: 12/31/2022]
Abstract
Bone fragility fractures are caused by low bone mass or impaired bone quality. Osteoblast/osteoclast coordination determines bone mass, but the factors that control bone quality are poorly understood. Osteocytes regulate osteoblast and osteoclast activity on bone surfaces but can also directly reorganize the bone matrix to improve bone quality through perilacunar/canalicular remodeling; however, the molecular mechanisms remain unclear. We previously found that deleting the transcriptional regulators Yes-associated protein (YAP) and transcriptional co-activator with PDZ-motif (TAZ) from osteoblast-lineage cells caused lethality in mice due to skeletal fragility. Here, we tested the hypothesis that YAP and TAZ regulate osteocyte-mediated bone remodeling by conditional ablation of both YAP and TAZ from mouse osteocytes using 8 kb-DMP1-Cre. Osteocyte-conditional YAP/TAZ deletion reduced bone mass and dysregulated matrix collagen content and organization, which together decreased bone mechanical properties. Further, YAP/TAZ deletion impaired osteocyte perilacunar/canalicular remodeling by reducing canalicular network density, length, and branching, as well as perilacunar flourochrome-labeled mineral deposition. Consistent with recent studies identifying TGF-β as a key inducer of osteocyte expression of matrix-remodeling enzymes, YAP/TAZ deletion in vivo decreased osteocyte expression of matrix proteases MMP13, MMP14, and CTSK. In vitro, pharmacologic inhibition of YAP/TAZ transcriptional activity in osteocyte-like cells abrogated TGF-β-induced matrix protease gene expression. Together, these data show that YAP and TAZ control bone matrix accrual, organization, and mechanical properties by regulating osteocyte-mediated bone remodeling. Elucidating the signaling pathways that control perilacunar/canalicular remodeling may enable future therapeutic targeting of bone quality to reverse skeletal fragility. © 2019 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Christopher D Kegelman
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA.,Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Jennifer C Coulombe
- Department of Mechanical Engineering, University of Colorado, Boulder, CO, USA
| | - Kelsey M Jordan
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA.,Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Daniel J Horan
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Ling Qin
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
| | - Alexander G Robling
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Virginia L Ferguson
- Department of Mechanical Engineering, University of Colorado, Boulder, CO, USA
| | - Teresita M Bellido
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Joel D Boerckel
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA.,Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
50
|
Abstract
The Hippo pathway was initially discovered in Drosophila melanogaster as a key regulator of tissue growth. It is an evolutionarily conserved signaling cascade regulating numerous biological processes, including cell growth and fate decision, organ size control, and regeneration. The core of the Hippo pathway in mammals consists of a kinase cascade, MST1/2 and LATS1/2, as well as downstream effectors, transcriptional coactivators YAP and TAZ. These core components of the Hippo pathway control transcriptional programs involved in cell proliferation, survival, mobility, stemness, and differentiation. The Hippo pathway is tightly regulated by both intrinsic and extrinsic signals, such as mechanical force, cell-cell contact, polarity, energy status, stress, and many diffusible hormonal factors, the majority of which act through G protein-coupled receptors. Here, we review the current understanding of molecular mechanisms by which signals regulate the Hippo pathway with an emphasis on mechanotransduction and the effects of this pathway on basic biology and human diseases.
Collapse
Affiliation(s)
- Shenghong Ma
- Department of Pharmacology and Moores Cancer Center, University of California, San Diego, La Jolla, California 92093, USA; , , ,
| | - Zhipeng Meng
- Department of Pharmacology and Moores Cancer Center, University of California, San Diego, La Jolla, California 92093, USA; , , ,
| | - Rui Chen
- Department of Pharmacology and Moores Cancer Center, University of California, San Diego, La Jolla, California 92093, USA; , , ,
| | - Kun-Liang Guan
- Department of Pharmacology and Moores Cancer Center, University of California, San Diego, La Jolla, California 92093, USA; , , ,
| |
Collapse
|