1
|
Spalding VA, Fellenstein BA, Ahodantin J, Jeyarajan AJ, Wang Y, Khan SK, Xu M, Lin W, Alatrakchi N, Su L, Chung RT, Salloum S. YAP mediates HIV-related liver fibrosis. JHEP Rep 2024; 6:101163. [PMID: 39524207 PMCID: PMC11544392 DOI: 10.1016/j.jhepr.2024.101163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 06/18/2024] [Accepted: 06/26/2024] [Indexed: 11/16/2024] Open
Abstract
Background & Aims HIV accelerates liver fibrosis attributable to multiple etiologies, including HCV, HBV, and steatotic liver disease. Evidence also suggests that HIV infection itself is associated with liver fibrogenesis. Recent studies have implicated Yes-associated protein 1 (YAP1) and the upstream lysophosphatidic acid (LPA)/PI3K/AKT pathway as critical regulators of hepatic fibrogenesis, and suggest a connection to HIV-related liver fibrosis. However, the relationship between YAP/PI3K/AKT pathway activation and HIV-related liver fibrosis remains uncertain. Methods qPCR, western blot, immunofluorescence, and ELISA (replicates n ≥3) were performed in an unbiased humanized mouse model (NRG-hu HSC mice, n = 6), the precision cut liver slice ex vivo model, and both traditional in vitro models as well as a 3D spheroid system. Results YAP target gene mRNA and protein levels (ANKRD, CTGF, CYR61) were upregulated across all models exposed to HIV. Humanized mice infected with HIV had significant increases in the percentage of YAP-positive nuclei (2.2-fold) and the percentage area of Sirius Red collagen staining (3.3-fold) compared to control mice. Serum concentrations of LPA were increased 5.8-fold in people living with HIV compared to healthy controls. Modulation of LPAR1, PI3K, and AKT by either inhibitors or small-interfering RNAs abrogated the fibrotic effects of HIV exposure and downregulated YAP target genes within cultured liver cells. Conclusions The LPAR/PI3K/AKT axis is vital for the activation of YAP and hepatic fibrogenesis due to HIV infection. This novel mechanistic insight suggests new pharmacologic targets for treatment of liver fibrosis in people living with HIV. Impact and implications There are currently no FDA-approved treatments for cirrhosis, while liver disease is the second leading cause of mortality among people living with HIV after AIDS. Increased lysophosphatidic acid concentrations and AKT activation after HIV infection found in recent work suggest that the Hippo pathway may be a key regulator of HIV-related fibrogenesis. By linking lysophosphatidic acid signaling, YAP activation, and HIV-related fibrogenesis, this mechanism presents a target for future research into therapeutic interventions for not only HIV but also other liver diseases, e.g. metabolic dysfunction- or alcohol-associated liver disease.
Collapse
Affiliation(s)
- Volney A. Spalding
- Liver Center and Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
| | - Brian A. Fellenstein
- Liver Center and Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
| | - James Ahodantin
- Lineberger Comprehensive Cancer Center and Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC USA
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Andre J. Jeyarajan
- Liver Center and Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
| | - Yongtao Wang
- Liver Center and Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
| | - Sanjoy K. Khan
- Liver Center and Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
| | - Min Xu
- Liver Center and Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
| | - Wenyu Lin
- Liver Center and Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
| | - Nadia Alatrakchi
- Liver Center and Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
| | - Lishan Su
- Lineberger Comprehensive Cancer Center and Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC USA
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Raymond T. Chung
- Liver Center and Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
| | - Shadi Salloum
- Liver Center and Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
| |
Collapse
|
2
|
Kim CL, Lim SB, Kim DH, Sim YE, Kang LJ, Park SJ, Kim H, Roh TH, Mo JS, Jeong HS. Regulation of Hippo-YAP signaling axis by Isoalantolactone suppresses tumor progression in cholangiocarcinoma. Transl Oncol 2024; 46:101971. [PMID: 38797019 PMCID: PMC11152753 DOI: 10.1016/j.tranon.2024.101971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 04/12/2024] [Accepted: 04/17/2024] [Indexed: 05/29/2024] Open
Abstract
Cholangiocarcinoma (CCA) is a devastating malignancy characterized by aggressive tumor growth and limited treatment options. Dysregulation of the Hippo signaling pathway and its downstream effector, Yes-associated protein (YAP), has been implicated in CCA development and progression. In this study, we investigated the effects of Isoalantolactone (IALT) on CCA cells to elucidate its effect on YAP activity and its potential clinical significance. Our findings demonstrate that IALT exerts cytotoxic effects, induces apoptosis, and modulates YAP signaling in SNU478 cells. We further confirmed the involvement of the canonical Hippo pathway by generating LATS1/LATS2 knockout cells, highlighting the dependence of IALT-mediated apoptosis and YAP phosphorylation on the Hippo-LATS signaling axis. In addition, IALT suppressed cell growth and migration, partially dependent on YAP-TEAD activity. These results provide insights into the therapeutic potential of targeting YAP in CCA and provide a rationale for developing of YAP-targeted therapies for this challenging malignancy.
Collapse
Affiliation(s)
- Cho-Long Kim
- Department of Biomedical Sciences, Graduate School, Ajou University School of Medicine, Suwon 16499, South Korea
| | - Su-Bin Lim
- Department of Biomedical Sciences, Graduate School, Ajou University School of Medicine, Suwon 16499, South Korea
| | - Dong Hyun Kim
- Department of Biomedical Sciences, Graduate School, Ajou University School of Medicine, Suwon 16499, South Korea
| | - Ye Eun Sim
- Department of Biomedical Sciences, Graduate School, Ajou University School of Medicine, Suwon 16499, South Korea
| | - Li-Jung Kang
- Three-Dimensional Immune System Imaging Core Facility, Ajou University, Suwon 16499, South Korea
| | - Su Jung Park
- Division of Applied Medicine, School of Korean Medicine, Pusan National University, Yangsan 50612, South Korea
| | - Hyungwoo Kim
- Division of Pharmacology, School of Korean Medicine, Pusan National University, Yangsan 50612, South Korea
| | - Tae Hoon Roh
- Department of Neurosurgery, Ajou University School of Medicine, Suwon 16499, South Korea
| | - Jung-Soon Mo
- Department of Biomedical Sciences, Graduate School, Ajou University School of Medicine, Suwon 16499, South Korea; Institute of Medical Science, Ajou University School of Medicine, Suwon 16499, South Korea.
| | - Han-Sol Jeong
- Division of Applied Medicine, School of Korean Medicine, Pusan National University, Yangsan 50612, South Korea.
| |
Collapse
|
3
|
Ampadu F, Awasthi V, Joshi AD. Role of Mitogen-Activated Protein Kinase Kinase Kinase Kinase 4 Signaling in Liver and Metabolic Diseases. J Pharmacol Exp Ther 2024; 390:233-239. [PMID: 38844365 PMCID: PMC11264251 DOI: 10.1124/jpet.124.002065] [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: 01/04/2024] [Revised: 05/17/2024] [Accepted: 05/28/2024] [Indexed: 06/09/2024] Open
Abstract
MAP4K4 is a serine/threonine protein kinase belonging to the germinal center kinase subgroup of sterile 20 protein family of kinases. MAP4K4 has been involved in regulating multiple biologic processes and a plethora of pathologies, including systemic inflammation, cardiovascular diseases, cancers, and metabolic and hepatic diseases. Recently, multiple reports have indicated the upregulation of MAP4K4 expression and signaling in hyperglycemia and liver diseases. This review provides an overview of our current knowledge of MAP4K4 structure and expression, as well as its regulation and signaling, specifically in metabolic and hepatic diseases. Reviewing these promising studies will enrich our understanding of MAP4K4 signaling pathways and, in the future, will help us design innovative therapeutic interventions against metabolic and liver diseases using MAP4K4 as a target. SIGNIFICANCE STATEMENT: Although most studies on the involvement of MAP4K4 in human pathologies are related to cancers, only recently its role in liver and other metabolic diseases is beginning to unravel. This mini review discusses recent advancements in MAP4K4 biology within the context of metabolic dysfunction and comprehensively characterizes MAP4K4 as a clinically relevant therapeutic target against liver and metabolic diseases.
Collapse
Affiliation(s)
- Felix Ampadu
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Vibhudutta Awasthi
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Aditya D Joshi
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| |
Collapse
|
4
|
Xiong R, Fleming E, Caldwell R, Vernon SD, Kozhaya L, Gunter C, Bateman L, Unutmaz D, Oh J. BioMapAI: Artificial Intelligence Multi-Omics Modeling of Myalgic Encephalomyelitis / Chronic Fatigue Syndrome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.24.600378. [PMID: 38979186 PMCID: PMC11230215 DOI: 10.1101/2024.06.24.600378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Chronic diseases like ME/CFS and long COVID exhibit high heterogeneity with multifactorial etiology and progression, complicating diagnosis and treatment. To address this, we developed BioMapAI, an explainable Deep Learning framework using the richest longitudinal multi-'omics dataset for ME/CFS to date. This dataset includes gut metagenomics, plasma metabolome, immune profiling, blood labs, and clinical symptoms. By connecting multi-'omics to asymptom matrix, BioMapAI identified both disease- and symptom-specific biomarkers, reconstructed symptoms, and achieved state-of-the-art precision in disease classification. We also created the first connectivity map of these 'omics in both healthy and disease states and revealed how microbiome-immune-metabolome crosstalk shifted from healthy to ME/CFS. Thus, we proposed several innovative mechanistic hypotheses for ME/CFS: Disrupted microbial functions - SCFA (butyrate), BCAA (amino acid), tryptophan, benzoate - lost connection with plasma lipids and bile acids, and activated inflammatory and mucosal immune cells (MAIT, γδT cells) with INFγ and GzA secretion. These abnormal dynamics are linked to key disease symptoms, including gastrointestinal issues, fatigue, and sleep problems.
Collapse
Affiliation(s)
- Ruoyun Xiong
- The Jackson Laboratory, Farmington, Connecticut, USA. 06032
- The University of Connecticut Health Center, Farmington, Connecticut, USA. 06030
| | | | - Ryan Caldwell
- The Jackson Laboratory, Farmington, Connecticut, USA. 06032
| | | | - Lina Kozhaya
- The Jackson Laboratory, Farmington, Connecticut, USA. 06032
| | - Courtney Gunter
- The Jackson Laboratory, Farmington, Connecticut, USA. 06032
- The University of Connecticut Health Center, Farmington, Connecticut, USA. 06030
| | | | - Derya Unutmaz
- The Jackson Laboratory, Farmington, Connecticut, USA. 06032
| | - Julia Oh
- The Jackson Laboratory, Farmington, Connecticut, USA. 06032
| |
Collapse
|
5
|
Zhang Y, Yan HJ, Wu J. The Tumor Immune Microenvironment plays a Key Role in Driving the Progression of Cholangiocarcinoma. Curr Cancer Drug Targets 2024; 24:681-700. [PMID: 38213139 DOI: 10.2174/0115680096267791231115101107] [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: 06/17/2023] [Revised: 10/09/2023] [Accepted: 10/13/2023] [Indexed: 01/13/2024]
Abstract
Cholangiocarcinoma (CCA) is an epithelial cancer distinguished by bile duct cell differentiation and is also a fibroproliferative tumor. It is characterized by a dense mesenchyme and a complex tumor immune microenvironment (TME). The TME comprises both cellular and non-cellular components. The celluar component includes CCA cells, immune cells and mesenchymal cells represented by the cancer-associated fibroblasts (CAFs), while the non-cellular component is represented by mesenchymal elements such as the extracellular matrix (ECM). Recent studies have demonstrated the important role of the TME in the development, progression, and treatment resistance of CCA. These cell-associated prognostic markers as well as intercellular connections, may serve as potential therapeutic targets and could inspire new treatment approaches for CCA in the future. This paper aims to summarize the current understanding of CCA's immune microenvironment, focusing on immune cells, mesenchymal cells, ECM, intercellular interactions, and metabolism within the microenvironment.
Collapse
Affiliation(s)
- Ye Zhang
- Department of Oncology, The Third Affiliated Hospital of Soochow University, 185 Juqian St, Changzhou, 213003, China
| | - Hai-Jiao Yan
- Department of Oncology, The Third Affiliated Hospital of Soochow University, 185 Juqian St, Changzhou, 213003, China
| | - Jun Wu
- Department of Oncology, The Third Affiliated Hospital of Soochow University, 185 Juqian St, Changzhou, 213003, China
| |
Collapse
|
6
|
Singh P, Kaur L, Ghose S, Varshney S, Jyothi V, Ghosh S, Kommineni P, Kv S, Scaria V, Sivasubbu S, Chandak GR, Sengupta S. Maternal-Periconceptional Vitamin B12 Deficiency in Wistar Rats Leads to Sex-Specific Programming for Cardiometabolic Disease Risk in the Next Generation. J Nutr 2023; 153:3382-3396. [PMID: 37660953 DOI: 10.1016/j.tjnut.2023.08.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 08/18/2023] [Accepted: 08/24/2023] [Indexed: 09/05/2023] Open
Abstract
BACKGROUND Maternal vitamin B12 deficiency plays a vital role in fetal programming, as corroborated by previous studies on murine models and longitudinal human cohorts. OBJECTIVES This study assessed the effects of diet-induced maternal vitamin B12 deficiency on F1 offspring in terms of cardiometabolic health and normalization of these effects by maternal-periconceptional vitamin B12 supplementation. METHODS A diet-induced maternal vitamin B12 deficient Wistar rat model was generated in which female rats were either fed a control AIN-76A diet (with 0.01 g/kg vitamin B12) or the same diet with vitamin B12 removed. Females from the vitamin B12-deficient group were mated with males on the control diet. A subset of vitamin B12-deficient females was repleted with vitamin B12 on day 1 of conception. The offspring in the F1 generation were assessed for changes in body composition, plasma biochemistry, and molecular changes in the liver. A multiomics approach was used to obtain a mechanistic insight into the changes in the offspring liver. RESULTS We showed that a 36% reduction in plasma vitamin B12 levels during pregnancy in F0 females can lead to continued vitamin B12 deficiency (60%-70% compared with control) in the F1 offspring and program them for cardiometabolic adversities. These adversities, such as high triglycerides and low high-density lipoprotein cholesterol, were seen only among F1 males but not females. DNA methylome analysis of the liver of F1 3-mo-old offspring highlights sexual dimorphism in the alteration of methylation status of genes critical to signaling processes. Proteomics and targeted metabolomics analysis confirm that sex-specific alterations occur through modulations in PPAR signaling and steroid hormone biosynthesis pathway. Repletion of deficient mothers with vitamin B12 at conception normalizes most of the molecular and biochemical changes. CONCLUSIONS Maternal vitamin B12 deficiency has a programming effect on the next generation and increases the risk for cardiometabolic syndrome in a sex-specific manner. Normalization of the molecular risk markers on vitamin B12 supplementation indicates a causal role.
Collapse
Affiliation(s)
- Praveen Singh
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Lovejeet Kaur
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India; Translational Health Science and Technology Institute, Faridabad, India
| | - Subhoshree Ghose
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Swati Varshney
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Vislavath Jyothi
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India
| | - Sourav Ghosh
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | | | - Shamsudheen Kv
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
| | - Vinod Scaria
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Sridhar Sivasubbu
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Giriraj Ratan Chandak
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India.
| | - Shantanu Sengupta
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India.
| |
Collapse
|
7
|
Wang T, Wang D, Sun Y, Zhuang T, Li X, Yang H, Zang Y, Liu Z, Yang P, Zhang C, Cui J, Fu M, Zhang S, Su P, Li Z, Zhu J, Ding Y. Regulation of the Hippo/YAP axis by CXCR7 in the tumorigenesis of gastric cancer. J Exp Clin Cancer Res 2023; 42:297. [PMID: 37950281 PMCID: PMC10636825 DOI: 10.1186/s13046-023-02870-3] [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: 06/20/2023] [Accepted: 10/18/2023] [Indexed: 11/12/2023] Open
Abstract
BACKGROUND The Hippo pathway is crucial in organ size control and tumorigenesis. Dysregulation of the Hippo/YAP axis is commonly observed in gastric cancer, while effective therapeutic targets for the Hippo/YAP axis are lacking. Identification of reliable drug targets and the underlying mechanisms that could inhibit the activity of the Hippo/YAP axis and gastric cancer progression is urgently needed. METHODS We used several gastric cancer cell lines and xenograft models and performed immunoblotting, qPCR, and in vivo studies to investigate the function of CXCR7 in gastric cancer progression. RESULTS In our current study, we demonstrate that the membrane receptor CXCR7 (C-X-C chemokine receptor 7) is an important modulator of the Hippo/YAP axis. The activation of CXCR7 could stimulate gastric cancer cell progression through the Hippo/YAP axis in vitro and in vivo, while pharmaceutical inhibition of CXCR7 via ACT-1004-1239 could block tumorigenesis in gastric cancer. Molecular studies revealed that the activation of CXCR7 could dephosphorylate YAP and facilitate YAP nuclear accumulation and transcriptional activation in gastric cancer. CXCR7 functions via G-protein Gαq/11 and Rho GTPase to activate YAP activity. Interestingly, ChIP assays showed that YAP could bind to the promoter region of CXCR7 and facilitate its gene transcription, which indicates that CXCR7 is both the upstream signalling and downstream target of the Hippo/YAP axis in gastric cancer. CONCLUSION In general, we identified a novel positive feedback loop between CXCR7 and the Hippo/YAP axis, and blockade of CXCR7 could be a plausible strategy for gastric cancer.
Collapse
Affiliation(s)
- Tianshi Wang
- Department of General Surgery, The Second Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, 250033, China
| | - Dehai Wang
- Department of General Surgery, The Second Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, 250033, China
| | - Yanan Sun
- Department of General Surgery, The Second Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, 250033, China
| | - Ting Zhuang
- Xinxiang Key Laboratory of Tumor Migration and Invasion Precision Medicine, School of Medical Technology, Xinxiang Medical University, Xinxiang, 453003, Henan Province, P. R. China
- Department of Thoracic Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Shandong Province, P. R. China
| | - Xin Li
- Xinxiang Key Laboratory of Tumor Migration and Invasion Precision Medicine, School of Medical Technology, Xinxiang Medical University, Xinxiang, 453003, Henan Province, P. R. China
| | - Huijie Yang
- Xinxiang Key Laboratory of Tumor Migration and Invasion Precision Medicine, School of Medical Technology, Xinxiang Medical University, Xinxiang, 453003, Henan Province, P. R. China
| | - Yifeng Zang
- Department of General Surgery, The Second Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, 250033, China
| | - Ziping Liu
- Department of General Surgery, The Second Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, 250033, China
| | - Penghe Yang
- Xinxiang Key Laboratory of Tumor Migration and Invasion Precision Medicine, School of Medical Technology, Xinxiang Medical University, Xinxiang, 453003, Henan Province, P. R. China
| | - Chenmiao Zhang
- Xinxiang Key Laboratory of Tumor Migration and Invasion Precision Medicine, School of Medical Technology, Xinxiang Medical University, Xinxiang, 453003, Henan Province, P. R. China
| | - Jiayao Cui
- Xinxiang Key Laboratory of Tumor Migration and Invasion Precision Medicine, School of Medical Technology, Xinxiang Medical University, Xinxiang, 453003, Henan Province, P. R. China
| | - Mingxi Fu
- Xinxiang Key Laboratory of Tumor Migration and Invasion Precision Medicine, School of Medical Technology, Xinxiang Medical University, Xinxiang, 453003, Henan Province, P. R. China
| | - Shuqing Zhang
- Xinxiang Key Laboratory of Tumor Migration and Invasion Precision Medicine, School of Medical Technology, Xinxiang Medical University, Xinxiang, 453003, Henan Province, P. R. China
| | - Peng Su
- Department of Pathology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250033, China.
| | - Zhongbo Li
- Xinxiang Key Laboratory of Tumor Migration and Invasion Precision Medicine, School of Medical Technology, Xinxiang Medical University, Xinxiang, 453003, Henan Province, P. R. China.
| | - Jian Zhu
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China.
| | - Yinlu Ding
- Department of General Surgery, The Second Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, 250033, China.
| |
Collapse
|
8
|
Tang D, Xu H, Du X. The role of non-canonical Hippo pathway in regulating immune homeostasis. Eur J Med Res 2023; 28:498. [PMID: 37941053 PMCID: PMC10631157 DOI: 10.1186/s40001-023-01484-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 10/30/2023] [Indexed: 11/10/2023] Open
Abstract
The Hippo pathway is a crucial signaling pathway that is highly conserved throughout evolution for the regulation of organ size and maintenance of tissue homeostasis. Initial studies have primarily focused on the canonical Hippo pathway, which governs organ development, tissue regeneration, and tumorigenesis. In recent years, extensive research has revealed that the non-canonical Hippo pathway, centered around Mst1/2 as its core molecule, plays a pivotal role in immune response and function by synergistically interacting with other signal transduction pathways. Consequently, the non-canonical Hippo pathway assumes significant importance in maintaining immune system homeostasis. This review concentrates on the research progress of the non-canonical Hippo pathway in regulating innate immune cell anti-infection responses, maintaining redox homeostasis, responding to microenvironmental stiffness, and T-cell differentiation.
Collapse
Affiliation(s)
- Dagang Tang
- Department of Orthopedics, Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400021, China
| | - Huan Xu
- Department of Ophtalmology, Daping Hospital, Army Medical University, Chongqing, 400012, China
| | - Xing Du
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, No.1 YouYi Road, Yuanjiagang, Yu Zhong District, Chongqing, 400016, China.
- Orthopedic Laboratory of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
9
|
Yang S, Guo LJ, Liang Y, He ZM, Luo J, Mu YD. ADCY6 is a potential prognostic biomarker and suppresses OTSCC progression via Hippo signaling pathway. Kaohsiung J Med Sci 2023; 39:978-988. [PMID: 37574908 DOI: 10.1002/kjm2.12725] [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: 04/08/2023] [Revised: 05/26/2023] [Accepted: 06/07/2023] [Indexed: 08/15/2023] Open
Abstract
Oral tongue squamous cell carcinoma (OTSCC) is a malignant tumor. Recently, studies have found that adenylate cyclase 6 (ADCY6) plays a pivotal role in many lethal tumors formation processes. The role of ADCY6 in OTSCC remains unknown. The expression of ADCY6 in OTSCC tissue samples was detected. The clinical significance of ADCY6 in OTSCC was analyzed by statistical methods. OTSCC cell lines were selected to analyze the biological function of ADCY6. Meanwhile, the effect of ADCY6 on the growth of OTSCC in vivo was explored using subcutaneous tumorigenesis assay. WB assay was used to detect the underlying signaling pathway. Cell function recovery test used to investigate the mechanism of ADCY6-promoting OTSCC malignant biological behavior via Hippo signaling pathway. We report that ADCY6 was obviously downregulated in OTSCC tissue samples and cell lines. Importantly, lower expression of ADCY6 indicates a poorer prognosis in patients with OTSCC, and its expression is significantly correlated with TNM stage and tumor size. Functionally, forced expression of ADCY6 can significantly inhibit the proliferation, migration, invasion, and promote apoptosis of OTSCC cells. Mechanistically, we demonstrated that ADCY6 upregulation impaired Hippo signaling pathway to reduce the malignant biological behavior of OTSCC. Generally, our findings suggest that ADCY6 suppressed Hippo signaling pathway to regulate malignant biological behavior in OTSCC, which provide new cues for further exploring the mechanism of occurrence and development of OTSCC.
Collapse
Affiliation(s)
- Sen Yang
- Department of Stomatology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Department of Oral and Maxillofacial Surgery, Suining Central Hospital, Sichuan, China
| | - Li-Juan Guo
- Department of Medical Cosmetology, Suining Central Hospital, Sichuan, China
| | - Yong Liang
- Institute of Electronic and Information Engineering of UESTC in Guangdong, University of Electronic Science and Technology of China, Dongguan, China
| | - Zhi-Ming He
- Institute of Electronic and Information Engineering of UESTC in Guangdong, University of Electronic Science and Technology of China, Dongguan, China
| | - Jia Luo
- Department of Stomatology Center, Suining Central Hospital, Sichuan, China
| | - Yan-Dong Mu
- Department of Stomatology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
10
|
Derham JM, Kalsotra A. The discovery, function, and regulation of epithelial splicing regulatory proteins (ESRP) 1 and 2. Biochem Soc Trans 2023; 51:1097-1109. [PMID: 37314029 PMCID: PMC11298080 DOI: 10.1042/bst20221124] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 06/01/2023] [Accepted: 06/05/2023] [Indexed: 06/15/2023]
Abstract
Alternative splicing is a broad and evolutionarily conserved mechanism to diversify gene expression and functionality. The process relies on RNA binding proteins (RBPs) to recognize and bind target sequences in pre-mRNAs, which allows for the inclusion or skipping of various alternative exons. One recently discovered family of RBPs is the epithelial splicing regulatory proteins (ESRP) 1 and 2. Here, we discuss the structure and physiological function of the ESRPs in a variety of contexts. We emphasize the current understanding of their splicing activities, using the classic example of fibroblast growth factor receptor 2 mutually exclusive splicing. We also describe the mechanistic roles of ESRPs in coordinating the splicing and functional output of key signaling pathways that support the maintenance of, or shift between, epithelial and mesenchymal cell states. In particular, we highlight their functions in the development of mammalian limbs, the inner ear, and craniofacial structure while discussing the genetic and biochemical evidence that showcases their conserved roles in tissue regeneration, disease, and cancer pathogenesis.
Collapse
Affiliation(s)
- Jessica M. Derham
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Auinash Kalsotra
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, IL, USA
- Cancer Center @ Illinois, University of Illinois Urbana-Champaign, Urbana, IL, USA
- Carl R. Woese Institute of Genomic Biology, University of Illinois Urbana-Champaign, Urbana, IL, USA
| |
Collapse
|
11
|
Zhao Y, Wang H, He T, Ma B, Chen G, Tzeng C. Knockdown of Yap attenuates TAA-induced hepatic fibrosis by interaction with hedgehog signals. J Cell Commun Signal 2023:10.1007/s12079-023-00775-6. [PMID: 37338798 DOI: 10.1007/s12079-023-00775-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 06/05/2023] [Indexed: 06/21/2023] Open
Abstract
Liver fibrosis is an aberrant wound healing response to tissue injury characterized by excessive extracellular matrix deposition and loss of normal liver architecture. Hepatic stellate cells (HSCs) activation is regards to be the major process in liver fibrogenesis which is dynamic and reversible. Both Hippo signaling core factor Yap and Hedgehog (Hh) signaling promote HSCs transdifferentiation thereby regulating the repair process of liver injury. However, the molecular function of YAP and the regulation between Yap and Hh during fibrogenesis remain uncertain. In this study, the essential roles of Yap in liver fibrosis were investigated. Yap was detected to be increased in liver fibrotic tissue by the thioacetamide (TAA)-induced zebrafish embryonic and adult models. Inhibition of Yap by both embryonic morpholino interference and adult's inhibitor treatment was proved to alleviate TAA-induced liver lesions by and histology and gene expression examination. Transcriptomic analysis and gene expression detection showed that Yap and Hh signaling pathway have a cross talking upon TAA-induced liver fibrosis. In addition, TAA induction promoted the nuclear colocalization of YAP and Hh signaling factor GLI2α. This study demonstrates that Yap and Hh play synergistic protective roles in liver fibrotic response and provides new theoretical insight concerning the mechanisms of fibrosis progression.
Collapse
Affiliation(s)
- Ye Zhao
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211800, China.
| | - Huiling Wang
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211800, China
| | - Tianhua He
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211800, China
| | - Bo Ma
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211800, China
| | - Guoguang Chen
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211800, China
| | - Chimeng Tzeng
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361005, China.
- Translational Medicine Research Center-Key Laboratory for Cancer T-Cell Theragnostic and Clinical Translation, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China.
- Xiamen Chang Gung Hospital Medical Research Center, Xiamen, Fujian, China.
| |
Collapse
|
12
|
Molecular insights of Hippo signaling in the chick developing lung. BIOCHIMICA ET BIOPHYSICA ACTA. GENE REGULATORY MECHANISMS 2023; 1866:194904. [PMID: 36572276 DOI: 10.1016/j.bbagrm.2022.194904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 12/18/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022]
Abstract
Hippo signaling pathway and its effector YAP have been recognized as an essential growth regulator during embryonic development. Hippo has been studied in different contexts; nevertheless, its role during chick lung branching morphogenesis remains unknown. Therefore, this work aims to determine Hippo role during early pulmonary organogenesis in the avian animal model. The current study describes the spatial distribution of Hippo signaling members in the embryonic chick lung by in situ hybridization. Overall, their expression is comparable to their mammalian counterparts. Moreover, the expression levels of phosphorylated-YAP (pYAP) and total YAP revealed that Hippo signaling is active in the embryonic chick lung. Furthermore, the presence of pYAP in the cytoplasm demonstrated that the Hippo machinery distribution is maintained in this tissue. In vitro studies were performed to assess the role of the Hippo signaling pathway in lung branching. Lung explants treated with a YAP/TEAD complex inhibitor (verteporfin) displayed a significant reduction in lung size and branching and decreased expression of ctgf (Hippo target gene) compared to the control. This approach also revealed that Hippo seems to modulate the expression of key molecular players involved in lung branching morphogenesis (sox2, sox9, axin2, and gli1). Conversely, when treated with dobutamine, an upstream regulator that promotes YAP phosphorylation, explant morphology was not severely affected. Overall, our data indicate that Hippo machinery is present and active in the early stages of avian pulmonary branching and that YAP is likely involved in the regulation of lung growth.
Collapse
|
13
|
Chu J, Li Y, He M, Zhang H, Yang L, Yang M, Liu J, Cui C, Hong L, Hu X, Zhou L, Li T, Li C, Fan H, Jiang G, Lang T. Zinc finger and SCAN domain containing 1, ZSCAN1, is a novel stemness-related tumor suppressor and transcriptional repressor in breast cancer targeting TAZ. Front Oncol 2023; 13:1041688. [PMID: 36923432 PMCID: PMC10009259 DOI: 10.3389/fonc.2023.1041688] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Accepted: 01/30/2023] [Indexed: 03/02/2023] Open
Abstract
Introduction Cancer stem cells (CSCs) targeted therapy holds the potential for improving cancer management; identification of stemness-related genes in CSCs is necessary for its development. Methods The Cancer Genome Atlas (TCGA) and the Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) datasets were used for survival analysis. ZSCAN1 correlated genes was identified by Spearman correlation analysis. Breast cancer stem-like cells (BCSLCs) were isolated by sorting CD44+CD24- cells from suspension cultured breast cancer (BC) spheroids. The sphere-forming capacity and sphere- and tumor-initiating capacities were determined by sphere formation and limiting dilution assays. The relative gene expression was determined by qRT-PCR, western blot. Lentivirus system was used for gene manipulation. Nuclear run-on assay was employed to examine the levels of nascent mRNAs. DNA pull-down and Chromatin immunoprecipitation (ChIP) assays were used for determining the interaction between protein and target DNA fragments. Luciferase reporter assay was used for evaluating the activity of the promoter. Results and discussion ZSCAN1 is aberrantly suppressed in BC, and this suppression indicates a bad prognosis. Ectopic expression of ZSCAN1 inhibited the proliferation, clonogenicity, and tumorigenicity of BC cells. ZSCAN1-overexpressing BCSLCs exhibited weakened stemness properties. Normal human mammary epithelial (HMLE) cells with ZSCAN1 depletion exhibited enhanced stemness properties. Mechanistic studies showed that ZSCAN1 directly binds to -951 ~ -925bp region of WWTR1 (encodes TAZ) promoter, inhibits WWTR1 transcription, thereby inhibiting the stemness of BCSCs. Our work thus revealed ZSCAN1 as a novel stemness-related tumor suppressor and transcriptional repressor in BC.
Collapse
Affiliation(s)
- Jian Chu
- Department of Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yunzhe Li
- College of Bioengineering, Chongqing University, Chongqing, China
| | - Misi He
- Department of Gynecologic Oncology, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, Chongqing, China
| | - Hui Zhang
- Department of Breast Cancer Center, Chongqing University Cancer Hospital, Chongqing, China
| | - Lingling Yang
- School of Medicine, Chongqing University, Chongqing, China
| | - Muyao Yang
- College of Bioengineering, Chongqing University, Chongqing, China
| | - Jingshu Liu
- Obstetrics and Gynecology Department, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chenxi Cui
- School of Medicine, Chongqing University, Chongqing, China
| | - Liquan Hong
- Department of Clinical Laboratory, Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Xingchi Hu
- Department of General Surgery, Yancheng City No.1 People's Hospital, Yancheng, Jiangsu, China
| | - Lei Zhou
- School of Optometry, Department of Applied Biology and Chemical Technology, Research Centre for SHARP Vision (RCSV), The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China.,Centre for Eye and Vision Research (CEVR), 17W Hong Kong Science Park, Hong Kong, Hong Kong SAR, China
| | - Tangya Li
- Department of General Surgery, Yancheng City No.1 People's Hospital, Yancheng, Jiangsu, China
| | - Changchun Li
- Department of General Surgery, Yancheng City No.1 People's Hospital, Yancheng, Jiangsu, China
| | - Huiwen Fan
- Department of General Surgery, Yancheng City No.1 People's Hospital, Yancheng, Jiangsu, China
| | - Guoqin Jiang
- Department of Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Tingyuan Lang
- Department of Gynecologic Oncology, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, Chongqing, China.,Reproductive Medicine Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
14
|
Pibiri M, Simbula G. Role of the Hippo pathway in liver regeneration and repair: recent advances. Inflamm Regen 2022; 42:59. [PMID: 36471376 PMCID: PMC9720992 DOI: 10.1186/s41232-022-00235-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 11/09/2022] [Indexed: 12/12/2022] Open
Abstract
Although the signaling pathways involved in normal liver regeneration have been well characterized, less has been done for livers affected by chronic tissue damage. These "abnormal livers" have an impaired regenerative response that leads to liver repair and fibrosis. The tumor suppressor Hippo pathway plays a key role in liver regeneration and repair. On this basis, this review discusses recent studies focusing on the involvement of the Hippo signaling pathway during "normal healthy liver regeneration" (i.e., in a normal liver after 2/3 partial hepatectomy) and "abnormal liver regeneration" (i.e., in a liver damaged by chronic disease). This could be an important question to address with respect to new therapies aimed at improving impaired liver regenerative responses. The studies reported here have shown that activation of the Hippo coactivators YAP/TAZ during normal liver regeneration promotes the formation of a new bile duct network through direct BEC proliferation or/and hepatocyte dedifferentiation to HPCs which can trans-differentiate to BECs. Moreover, YAP/TAZ signaling interaction with other signaling pathways mediates the recruitment and activation of Kupffer cells, which release mitogenic cytokines for parenchymal and/or non-parenchymal cells and engage in phagocytosis of cellular debris. In addition, YAP-mediated activation of stellate cells (HSCs) promotes liver regeneration through the synthesis of extracellular matrix. However, in chronically diseased livers, where the predetermined threshold for proper liver regeneration is exceeded, YAP/TAZ activation results in a reparative process characterized by liver fibrosis. In this condition, YAP/TAZ activation in parenchymal and non-parenchymal cells results in (i) differentiation of quiescent HSCs into myofibroblastic HSCs; (ii) recruitment of macrophages releasing inflammatory cytokines; (iii) polarization of macrophages toward the M2 phenotype. Since accumulation of damaged hepatocytes in chronic liver injury represent a significant risk factor for the development of hepatocarcinoma, this review also discussed the involvement of the Hippo pathway in the clearance of damaged cells.
Collapse
Affiliation(s)
- Monica Pibiri
- grid.7763.50000 0004 1755 3242Department of Biomedical Sciences, Oncology and Molecular Pathology Unit, University of Cagliari, Cittadella Universitaria di Monserrato, S.P. Monserrato-Sestu km 0.700, Blocco A. 09042 Monserrato, Cagliari, Italy
| | - Gabriella Simbula
- grid.7763.50000 0004 1755 3242Department of Biomedical Sciences, Oncology and Molecular Pathology Unit, University of Cagliari, Cittadella Universitaria di Monserrato, S.P. Monserrato-Sestu km 0.700, Blocco A. 09042 Monserrato, Cagliari, Italy
| |
Collapse
|
15
|
Yang T, Wu E, Zhu X, Leng Y, Ye S, Dong R, Liu J, Zhong J, Zheng Y, Xu W, Luo J, Kong L, Zhang H. TKF, a mexicanolide-type limonoid derivative, suppressed hepatic stellate cells activation and liver fibrosis through inhibition of the YAP/Notch3 pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 107:154466. [PMID: 36182796 DOI: 10.1016/j.phymed.2022.154466] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 08/02/2022] [Accepted: 09/18/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND Liver fibrosis is a common scarring response and may ultimately lead to liver cancer, unfortunately, there is currently no effective antifibrotic drug approved for human use. Limonoids exhibit a broad spectrum of biological activities; however, the potential role of limonoids against fibrosis is largely unknown. PURPOSE This study investigates the antifibrotic activities and potential mechanisms of TKF (3-tigloyl-khasenegasin F), a natural mexicanolide-type limonoid derivative. STUDY DESIGN/METHODS Two well-established mouse models (CCl4 challenge and bile duct ligation) were used to assess anti-fibrotic effects of TKF in vivo. Human hepatic stellate cell (HSC) line LX-2 and mouse primary hepatic stellate cells (pHSCs) also served as in vitro liver fibrosis models. RESULT TKF administration significantly attenuated hepatic histopathological injury and collagen accumulation and suppressed fibrogenesis-associated gene expression including Col1a1, Acta2, and Timp1. In LX-2 cells and mouse pHSCs, TKF dose-dependently suppressed HSC activation and the expression levels of fibrogenic markers. Mechanistic studies showed that TKF inhibited Notch3-Hes1 and YAP signalings in vivo and in vitro. Furthermore, YAP inhibition or knockdown downregulated the Notch3 expression; however, Notch3 inhibition or knockdown did not affect the level of YAP in activated HSC. We revealed that TKF inhibited Notch3-Hes1 activation and downregulated hepatic fibrogenic gene expression via inhibiting YAP. CONCLUSION The therapeutic benefit of TKF against liver fibrosis results from inhibition of YAP and Notch3-Hes1 pathways, indicating that TKF may be a novel therapeutic candidate for liver fibrosis.
Collapse
Affiliation(s)
- Ting Yang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Enyi Wu
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xiaoyun Zhu
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yingrong Leng
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Shengtao Ye
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Ruirui Dong
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Jiaman Liu
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Jiawen Zhong
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Ying Zheng
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Wenjun Xu
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Jun Luo
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Lingyi Kong
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China.
| | - Hao Zhang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
16
|
Zhang Y, Xu H, Cui G, Liang B, Chen X, Ko S, Affo S, Song X, Liao Y, Feng J, Wang P, Wang H, Xu M, Wang J, Pes GM, Ribback S, Zeng Y, Singhi A, Schwabe RF, Monga SP, Evert M, Tang L, Calvisi DF, Chen X. β-Catenin Sustains and Is Required for YES-associated Protein Oncogenic Activity in Cholangiocarcinoma. Gastroenterology 2022; 163:481-494. [PMID: 35489428 PMCID: PMC9329198 DOI: 10.1053/j.gastro.2022.04.028] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 04/05/2022] [Accepted: 04/19/2022] [Indexed: 02/08/2023]
Abstract
BACKGROUND & AIMS YES-associated protein (YAP) aberrant activation is implicated in intrahepatic cholangiocarcinoma (iCCA). Transcriptional enhanced associate domain (TEAD)-mediated transcriptional regulation is the primary signaling event downstream of YAP. The role of Wnt/β-Catenin signaling in cholangiocarcinogenesis remains undetermined. Here, we investigated the possible molecular interplay between YAP and β-Catenin cascades in iCCA. METHODS Activated AKT (Myr-Akt) was coexpressed with YAP (YapS127A) or Tead2VP16 via hydrodynamic tail vein injection into mouse livers. Tumor growth was monitored, and liver tissues were collected and analyzed using histopathologic and molecular analysis. YAP, β-Catenin, and TEAD interaction in iCCAs was investigated through coimmunoprecipitation. Conditional Ctnnb1 knockout mice were used to determine β-Catenin function in murine iCCA models. RNA sequencing was performed to analyze the genes regulated by YAP and/or β-Catenin. Immunostaining of total and nonphosphorylated/activated β-Catenin staining was performed in mouse and human iCCAs. RESULTS We discovered that TEAD factors are required for YAP-dependent iCCA development. However, transcriptional activation of TEADs did not fully recapitulate YAP's activities in promoting cholangiocarcinogenesis. Notably, β-Catenin physically interacted with YAP in human and mouse iCCA. Ctnnb1 ablation strongly suppressed human iCCA cell growth and Yap-dependent cholangiocarcinogenesis. Furthermore, RNA-sequencing analysis revealed that YAP/ transcriptional coactivator with PDZ-binding motif (TAZ) regulate a set of genes significantly overlapping with those controlled by β-Catenin. Importantly, activated/nonphosphorylated β-Catenin was detected in more than 80% of human iCCAs. CONCLUSION YAP induces cholangiocarcinogenesis via TEAD-dependent transcriptional activation and interaction with β-Catenin. β-Catenin binds to YAP in iCCA and is required for YAP full transcriptional activity, revealing the functional crosstalk between YAP and β-Catenin pathways in cholangiocarcinogenesis.
Collapse
Affiliation(s)
- Yi Zhang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China; Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, California
| | - Hongwei Xu
- Department of Liver Surgery, Center of Liver Transplantation, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Guofei Cui
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, California
| | - Binyong Liang
- Hepatic Surgery Center, Department of Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiangzheng Chen
- Liver Transplantation Division, Department of Liver Surgery, and Laboratory of Liver Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Sungjin Ko
- Department of Pathology and Medicine, and Pittsburgh Liver Research Center, University of Pittsburgh School of Medicine, and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Silvia Affo
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Xinhua Song
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Yi Liao
- The Central Laboratory, Shenzhen Second People's Hospital/First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, Guangdong, China
| | - Jianguo Feng
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China; Laboratory of Anesthesiology, Southwest Medical University, Luzhou, China
| | - Pan Wang
- Collaborative Innovation Center for Agricultural Product Processing and Nutrition & Health, Beijing Vegetable Research Center, Beijing Academy of Agriculture and Forestry Science, Beijing, China
| | - Haichuan Wang
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, California; Liver Transplantation Division, Department of Liver Surgery, and Laboratory of Liver Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Meng Xu
- Department of General Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an, China
| | - Jingxiao Wang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Giovanni M Pes
- Department of Medical, Surgical, and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Silvia Ribback
- Institute of Pathology, University of Greifswald, Greifswald, Germany
| | - Yong Zeng
- Liver Transplantation Division, Department of Liver Surgery, and Laboratory of Liver Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Aatur Singhi
- Department of Pathology and Medicine, and Pittsburgh Liver Research Center, University of Pittsburgh School of Medicine, and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | | | - Satdarshan P Monga
- Department of Pathology and Medicine, and Pittsburgh Liver Research Center, University of Pittsburgh School of Medicine, and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Matthias Evert
- Institute of Pathology, University of Regensburg, Regensburg, Germany
| | - Liling Tang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China.
| | - Diego F Calvisi
- Institute of Pathology, University of Regensburg, Regensburg, Germany.
| | - Xin Chen
- Department of Bioengineering and Therapeutic Sciences and Liver Center, University of California, San Francisco, California; Cancer Biology Program, University of Hawaii Cancer Center, Honolulu, Hawaii.
| |
Collapse
|
17
|
Zhang YY, Hu ZL, Qi YH, Li HY, Chang X, Gao XX, Liu CH, Li YY, Lou JH, Zhai Y, Li CQ. Pretreatment of nucleus pulposus mesenchymal stem cells with appropriate concentration of H 2O 2 enhances their ability to treat intervertebral disc degeneration. Stem Cell Res Ther 2022; 13:340. [PMID: 35883157 PMCID: PMC9327256 DOI: 10.1186/s13287-022-03031-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 07/04/2022] [Indexed: 11/18/2022] Open
Abstract
Background Nucleus pulposus mesenchymal stem cells (NPMSCs) transplantation is a promising treatment for intervertebral disc degeneration (IVDD). However, the transplanted NPMSCs exhibited weak cell proliferation, high cell apoptosis, and a low ability to resist the harsh microenvironment of the degenerated intervertebral disc. There is an urgent need to explore feasible methods to enhance the therapeutic efficacy of NPMSCs transplantation. Objective To identify the optimal concentration for NPMSCs pretreatment with hydrogen peroxide (H2O2) and explore the therapeutic efficacy of NPMSCs transplantation using H2O2 pretreatment in IVDD. Methods Rat NPMSCs were pretreated with different concentrations (range from 25 to 300 μM) of H2O2. The proliferation, reactive oxygen species (ROS) level, and apoptosis of NPMSCs were detected by cell counting kit-8 (CCK-8) assay, 5-ethynyl-2′-deoxyuridine (EdU) staining, and flow cytometry in vitro. The underlying signalling pathways were explored utilizing Western blotting. A rat needle puncture-stimulated IVDD model was established. X-ray, histological staining, and a multimode small animal live imaging system were used to evaluate the therapeutic effect of H2O2-pretreated NPMSCs in vivo. Results NPMSCs pretreated with 75 μM H2O2 demonstrated the strongest elevated cell proliferation by inhibiting the Hippo pathway (P < 0.01). Meanwhile, 75 μM H2O2-pretreated NPMSCs exhibited significantly enhanced antioxidative stress ability (P < 0.01), which is related to downregulated Brd4 and Keap1 and upregulated Nrf2. NPMSCs pretreated with 75 μM H2O2 also exhibited distinctly decreased apoptosis (P < 0.01). In vivo experiments verified that 75 μM H2O2-pretreated NPMSCs-transplanted rats exhibited an enhanced disc height index (DHI% = 90.00 ± 4.55, P < 0.01) and better histological morphology (histological score = 13.5 ± 0.5, P < 0.01), which means 75 μM H2O2-pretreated NPMSCs can better adapt to the environment of degenerative intervertebral discs and promote the repair of IVDD. Conclusions Pretreatment with 75 μM H2O2 was the optimal concentration to improve the proliferation, antioxidative stress, and antiapoptotic ability of transplanted NPMSCs, which is expected to provide a new feasible method to improve the stem cell therapy efficacy of IVDD. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-03031-7.
Collapse
Affiliation(s)
- Yu-Yao Zhang
- Department of Orthopedics, Xinqiao Hospital, Army Military Medical University, Chongqing, 400037, China
| | - Zhi-Lei Hu
- Department of Orthopedics, Xinqiao Hospital, Army Military Medical University, Chongqing, 400037, China
| | - Yu-Han Qi
- Institute of Basic Theory of Traditional Chinese Medicine, China Academy of Chinese Medical Science, Beijing, 100000, China
| | - Hai-Yin Li
- Department of Orthopedics, Xinqiao Hospital, Army Military Medical University, Chongqing, 400037, China
| | - Xian Chang
- Department of Orthopedics, Xinqiao Hospital, Army Military Medical University, Chongqing, 400037, China
| | - Xiao-Xin Gao
- Department of Orthopedics, Xinqiao Hospital, Army Military Medical University, Chongqing, 400037, China
| | - Chen-Hao Liu
- Department of Orthopedics, Xinqiao Hospital, Army Military Medical University, Chongqing, 400037, China
| | - Yue-Yang Li
- Department of Orthopedics, Xinqiao Hospital, Army Military Medical University, Chongqing, 400037, China
| | - Jin-Hui Lou
- Department of Orthopedics, Xinqiao Hospital, Army Military Medical University, Chongqing, 400037, China
| | - Yu Zhai
- Department of Orthopedics, Xinqiao Hospital, Army Military Medical University, Chongqing, 400037, China.
| | - Chang-Qing Li
- Department of Orthopedics, Xinqiao Hospital, Army Military Medical University, Chongqing, 400037, China.
| |
Collapse
|
18
|
Arora T, Kausar MA, Aboelnaga SM, Anwar S, Hussain MA, Sadaf S, Kaur S, Eisa AA, Shingatgeri VMM, Najm MZ, Aloliqi AA. miRNAs and the Hippo pathway in cancer: Exploring the therapeutic potential (Review). Oncol Rep 2022; 48:135. [PMID: 35699111 DOI: 10.3892/or.2022.8346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 05/17/2022] [Indexed: 11/06/2022] Open
Abstract
Cancer is recognized as the leading cause of death worldwide. The hippo signaling pathway regulates organ size by balancing cell proliferation and cell death; hence dysregulation of the hippo pathway promotes cancer‑like conditions. miRNAs are a type of non‑coding RNA that have been shown to regulate gene expression. miRNA levels are altered in various classes of cancer. Researchers have also uncovered a crosslinking between miRNAs and the hippo pathway, which has been linked to cancer. The components of the hippo pathway regulate miRNA synthesis, and various miRNAs regulate the components of the hippo pathway both positively and negatively, which can lead to cancer‑like conditions. In the present review article, the mechanism behind the hippo signaling pathway and miRNAs biogenesis and crosslinks between miRNAs and the hippo pathway, which result in cancer, shall be discussed. Furthermore, the article will cover miRNA‑related therapeutics and provide an overview of the development of resistance to anticancer drugs. Understanding the underlying processes would improve the chances of developing effective cancer treatment therapies.
Collapse
Affiliation(s)
- Taruna Arora
- Division of Reproductive Biology, Maternal & Child Health, Department of Health Research, ICMR, MOHFW, Government of India, Ansari Nagar, New Delhi 110029, India
| | - Mohd Adnan Kausar
- Department of Biochemistry, College of Medicine, University of Hail, Hail, KSA‑2240, Saudi Arabia
| | | | - Sadaf Anwar
- Department of Biochemistry, College of Medicine, University of Hail, Hail, KSA‑2240, Saudi Arabia
| | - Malik Asif Hussain
- Department of Pathology, University of Hail, Hail, KSA-2240, Saudi Arabia
| | - Sadaf Sadaf
- Department of Biotechnology, Jamia Millia Islamia, New Delhi 110025, India
| | - Simran Kaur
- School of Biosciences, Apeejay Stya University, Sohna, Haryana 122103, India
| | - Alaa Abdulaziz Eisa
- Department of Medical Laboratories Technology, College of Applied Medical Sciences, Taibah University, Medina, KSA‑344, Saudi Arabia
| | | | | | - Abdulaziz A Aloliqi
- Department of Medical Biotechnology, College of Applied Medical Sciences, Qassim University, Buraydah 51542, Saudi Arabia
| |
Collapse
|
19
|
López-Mengual A, Segura-Feliu M, Sunyer R, Sanz-Fraile H, Otero J, Mesquida-Veny F, Gil V, Hervera A, Ferrer I, Soriano J, Trepat X, Farré R, Navajas D, Del Río JA. Involvement of Mechanical Cues in the Migration of Cajal-Retzius Cells in the Marginal Zone During Neocortical Development. Front Cell Dev Biol 2022; 10:886110. [PMID: 35652101 PMCID: PMC9150848 DOI: 10.3389/fcell.2022.886110] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/25/2022] [Indexed: 12/24/2022] Open
Abstract
Emerging evidence points to coordinated action of chemical and mechanical cues during brain development. At early stages of neocortical development, angiogenic factors and chemokines such as CXCL12, ephrins, and semaphorins assume crucial roles in orchestrating neuronal migration and axon elongation of postmitotic neurons. Here we explore the intrinsic mechanical properties of the developing marginal zone of the pallium in the migratory pathways and brain distribution of the pioneer Cajal-Retzius cells. These neurons are generated in several proliferative regions in the developing brain (e.g., the cortical hem and the pallial subpallial boundary) and migrate tangentially in the preplate/marginal zone covering the upper portion of the developing cortex. These cells play crucial roles in correct neocortical layer formation by secreting several molecules such as Reelin. Our results indicate that the motogenic properties of Cajal-Retzius cells and their perinatal distribution in the marginal zone are modulated by both chemical and mechanical factors, by the specific mechanical properties of Cajal-Retzius cells, and by the differential stiffness of the migratory routes. Indeed, cells originating in the cortical hem display higher migratory capacities than those generated in the pallial subpallial boundary which may be involved in the differential distribution of these cells in the dorsal-lateral axis in the developing marginal zone.
Collapse
Affiliation(s)
- Ana López-Mengual
- Molecular and Cellular Neurobiotechnology, Institute for Bioengineering of Catalonia (IBEC), Barcelona, Spain.,Department of Cell Biology, Physiology and Immunology, Universitat de Barcelona, Barcelona, Spain.,Network Centre of Biomedical Research of Neurodegenerative Diseases (CIBERNED), Institute of Health Carlos III, Madrid, Spain.,Institute of Neuroscience, University of Barcelona, Barcelona, Spain
| | - Miriam Segura-Feliu
- Molecular and Cellular Neurobiotechnology, Institute for Bioengineering of Catalonia (IBEC), Barcelona, Spain.,Department of Cell Biology, Physiology and Immunology, Universitat de Barcelona, Barcelona, Spain.,Network Centre of Biomedical Research of Neurodegenerative Diseases (CIBERNED), Institute of Health Carlos III, Madrid, Spain.,Institute of Neuroscience, University of Barcelona, Barcelona, Spain
| | - Raimon Sunyer
- Unitat de Biofísica I Bioenginyeria, Universitat de Barcelona, Barcelona, Spain
| | - Héctor Sanz-Fraile
- Unitat de Biofísica I Bioenginyeria, Universitat de Barcelona, Barcelona, Spain
| | - Jorge Otero
- Unitat de Biofísica I Bioenginyeria, Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red en Enfermedades Respiratorias, Madrid, Spain
| | - Francina Mesquida-Veny
- Molecular and Cellular Neurobiotechnology, Institute for Bioengineering of Catalonia (IBEC), Barcelona, Spain.,Department of Cell Biology, Physiology and Immunology, Universitat de Barcelona, Barcelona, Spain.,Network Centre of Biomedical Research of Neurodegenerative Diseases (CIBERNED), Institute of Health Carlos III, Madrid, Spain.,Institute of Neuroscience, University of Barcelona, Barcelona, Spain
| | - Vanessa Gil
- Molecular and Cellular Neurobiotechnology, Institute for Bioengineering of Catalonia (IBEC), Barcelona, Spain.,Department of Cell Biology, Physiology and Immunology, Universitat de Barcelona, Barcelona, Spain.,Network Centre of Biomedical Research of Neurodegenerative Diseases (CIBERNED), Institute of Health Carlos III, Madrid, Spain.,Institute of Neuroscience, University of Barcelona, Barcelona, Spain
| | - Arnau Hervera
- Molecular and Cellular Neurobiotechnology, Institute for Bioengineering of Catalonia (IBEC), Barcelona, Spain.,Department of Cell Biology, Physiology and Immunology, Universitat de Barcelona, Barcelona, Spain.,Network Centre of Biomedical Research of Neurodegenerative Diseases (CIBERNED), Institute of Health Carlos III, Madrid, Spain.,Institute of Neuroscience, University of Barcelona, Barcelona, Spain
| | - Isidre Ferrer
- Institute of Neuroscience, University of Barcelona, Barcelona, Spain.,Senior Consultant, Bellvitge University Hospital, Hospitalet de Llobregat, Barcelona, Spain.,Department of Pathology and Experimental Therapeutics, University of Barcelona, Barcelona, Spain
| | - Jordi Soriano
- Departament de Física de La Matèria Condensada, Universitat de Barcelona, Barcelona, Spain.,University of Barcelona Institute of Complex Systems (UBICS), Barcelona, Spain
| | - Xavier Trepat
- Unitat de Biofísica I Bioenginyeria, Universitat de Barcelona, Barcelona, Spain.,Integrative Cell and Tissue Dynamics, Institute for Bioengineering of Catalonia (IBEC), Parc Científic de Barcelona, Barcelona, Spain.,Center for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain.,Institució Catalana de Recerca I Estudis Avançats, University of Barcelona, Barcelona, Spain
| | - Ramon Farré
- Unitat de Biofísica I Bioenginyeria, Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red en Enfermedades Respiratorias, Madrid, Spain.,Institut D'Investigacions Biomèdiques August Pi Sunyer, Barcelona, Spain
| | - Daniel Navajas
- Unitat de Biofísica I Bioenginyeria, Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red en Enfermedades Respiratorias, Madrid, Spain.,Cellular and Respiratory Biomechanics, Institute for Bioengineering of Catalonia (IBEC), Barcelona, Spain
| | - José Antonio Del Río
- Molecular and Cellular Neurobiotechnology, Institute for Bioengineering of Catalonia (IBEC), Barcelona, Spain.,Department of Cell Biology, Physiology and Immunology, Universitat de Barcelona, Barcelona, Spain.,Network Centre of Biomedical Research of Neurodegenerative Diseases (CIBERNED), Institute of Health Carlos III, Madrid, Spain.,Institute of Neuroscience, University of Barcelona, Barcelona, Spain
| |
Collapse
|
20
|
One of the Primary Functions of Tissue-Resident Pluripotent Pericytes Cells May Be to Regulate Normal Organ Growth and Maturation: Implications for Attempts to Repair Tissues Later in Life. Int J Mol Sci 2022; 23:ijms23105496. [PMID: 35628309 PMCID: PMC9146368 DOI: 10.3390/ijms23105496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 05/10/2022] [Accepted: 05/12/2022] [Indexed: 12/04/2022] Open
Abstract
Adult mesenchymal stem cells were reported more than 30 years ago. Since then, their potential to repair and regenerate damaged or diseased tissues has been studied intensively in both preclinical models and human trials. Most of the need for such tissue repair/regeneration is in older populations, so much of the effort has been performed with autologous cells in older patients. However, success has been difficult to achieve. In the literature, it has been noted that such progenitor cells from younger individuals often behave with more vigorous activity and are functionally enhanced compared to those from older individuals or animals. In addition, cells with the characteristics of mesenchymal stem cells or pluripotent mesenchymal regulatory cells exist in nearly all tissues and organs as pericytes since fetal life. Such evidence raises the possibility that one of the primary roles of these organ-specific cells is to regulate organ growth and maturation, and then subsequently play a role in the maintenance of organ integrity. This review will discuss the evidence to support this concept and the implications of such a concept regarding the use of these progenitor cells for the repair and regeneration of tissues damaged by injury or disease later in life. For the latter, it may be necessary to return the organ-specific progenitor cells to the functional state that contributed to their effectiveness during growth and maturation rather than attempting to use them after alterations imposed during the aging process have been established and their function compromised.
Collapse
|
21
|
Hsu SC, Lin CY, Lin YY, Collins CC, Chen CL, Kung HJ. TEAD4 as an Oncogene and a Mitochondrial Modulator. Front Cell Dev Biol 2022; 10:890419. [PMID: 35602596 PMCID: PMC9117765 DOI: 10.3389/fcell.2022.890419] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 04/18/2022] [Indexed: 11/13/2022] Open
Abstract
TEAD4 (TEA Domain Transcription Factor 4) is well recognized as the DNA-anchor protein of YAP transcription complex, which is modulated by Hippo, a highly conserved pathway in Metazoa that controls organ size through regulating cell proliferation and apoptosis. To acquire full transcriptional activity, TEAD4 requires co-activator, YAP (Yes-associated protein) or its homolog TAZ (transcriptional coactivator with PDZ-binding motif) the signaling hub that relays the extracellular stimuli to the transcription of target genes. Growing evidence suggests that TEAD4 also exerts its function in a YAP-independent manner through other signal pathways. Although TEAD4 plays an essential role in determining that differentiation fate of the blastocyst, it also promotes tumorigenesis by enhancing metastasis, cancer stemness, and drug resistance. Upregulation of TEAD4 has been reported in several cancers, including colon cancer, gastric cancer, breast cancer, and prostate cancer and serves as a valuable prognostic marker. Recent studies show that TEAD4, but not other members of the TEAD family, engages in regulating mitochondrial dynamics and cell metabolism by modulating the expression of mitochondrial- and nuclear-encoded electron transport chain genes. TEAD4’s functions including oncogenic activities are tightly controlled by its subcellular localization. As a predominantly nuclear protein, its cytoplasmic translocation is triggered by several signals, such as osmotic stress, cell confluency, and arginine availability. Intriguingly, TEAD4 is also localized in mitochondria, although the translocation mechanism remains unclear. In this report, we describe the current understanding of TEAD4 as an oncogene, epigenetic regulator and mitochondrial modulator. The contributing mechanisms will be discussed.
Collapse
Affiliation(s)
- Sheng-Chieh Hsu
- Program for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Ching-Yu Lin
- Program for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Yen-Yi Lin
- Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Colin C. Collins
- Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Chia-Lin Chen
- Program for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- Vancouver Prostate Centre and Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
- *Correspondence: Chia-Lin Chen, ; Hsing-Jien Kung,
| | - Hsing-Jien Kung
- Program for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Taiwan
- Department of Biochemistry and Molecular Medicine, Comprehensive Cancer Center, University of California, Davis, Sacramento, CA, United States
- *Correspondence: Chia-Lin Chen, ; Hsing-Jien Kung,
| |
Collapse
|
22
|
Target Therapy for Hepatocellular Carcinoma: Beyond Receptor Tyrosine Kinase Inhibitors and Immune Checkpoint Inhibitors. BIOLOGY 2022; 11:biology11040585. [PMID: 35453784 PMCID: PMC9027240 DOI: 10.3390/biology11040585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/19/2022] [Accepted: 04/07/2022] [Indexed: 11/24/2022]
Abstract
Simple Summary Hepatocellular carcinoma (HCC) is the most common type of primary liver cancer and its incidence is steadily increasing. The development of HCC is a complex, multi-step process that is accompanied by alterations in multiple signaling cascades. Recent years have seen advancement in understanding molecular signaling pathways that play central roles in hepatocarcinogenesis. Aberrant activation of YAP/TAZ, Hedgehog, or Wnt/β-catenin signaling is frequently found in a subset of HCC patients. Targeting the signaling pathway via small molecule inhibitors could be a promising therapeutic option for the subset of patients. In this review, we will introduce the signaling pathways, discuss their roles in the development of HCC, and propose a therapeutic approach targeting the signaling pathways in the context of HCC. Abstract Hepatocellular carcinoma (HCC) is a major health concern worldwide, and its incidence is increasing steadily. To date, receptor tyrosine kinases (RTKs) are the most favored molecular targets for the treatment of HCC, followed by immune checkpoint regulators such as PD-1, PD-L1, and CTLA-4. With less than desirable clinical outcomes from RTK inhibitors as well as immune checkpoint inhibitors (ICI) so far, novel molecular target therapies have been proposed for HCC. In this review, we will introduce diverse molecular signaling pathways that are aberrantly activated in HCC, focusing on YAP/TAZ, Hedgehog, and Wnt/β-catenin signaling pathways, and discuss potential therapeutic strategies targeting the signaling pathways in HCC.
Collapse
|
23
|
Simino LADP, Fontana MF, de Fante T, Panzarin C, Ignacio-Souza LM, Milanski M, Torsoni MA, Desai M, Ross MG, Torsoni AS. Hepatic Epigenetic Reprogramming After Liver Resection in Offspring Alleviates the Effects of Maternal Obesity. Front Cell Dev Biol 2022; 10:830009. [PMID: 35433669 PMCID: PMC9009519 DOI: 10.3389/fcell.2022.830009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 03/07/2022] [Indexed: 11/13/2022] Open
Abstract
Obesity has become a public health problem in recent decades, and during pregnancy, it can lead to an increased risk of gestational complications and permanent changes in the offspring resulting from a process known as metabolic programming. The offspring of obese dams are at increased risk of developing non-alcoholic fatty liver disease (NAFLD), even in the absence of high-fat diet consumption. NAFLD is a chronic fatty liver disease that can progress to extremely severe conditions that require surgical intervention with the removal of the injured tissue. Liver regeneration is necessary to preserve organ function. A range of pathways is activated in the liver regeneration process, including the Hippo, TGFβ, and AMPK signaling pathways that are under epigenetic control. We investigated whether microRNA modulation in the liver of the offspring of obese dams would impact gene expression of Hippo, TGFβ, and AMPK pathways and tissue regeneration after partial hepatectomy (PHx). Female Swiss mice fed a standard chow or a high-fat diet (HFD) before and during pregnancy and lactation were mated with male control mice. The offspring from control (CT-O) and obese (HF-O) dams weaned to standard chow diet until day 56 were submitted to PHx surgery. Prior to the surgery, HF-O presented alterations in miR-122, miR-370, and Let-7a expression in the liver compared to CT-O, as previously shown, as well as in its target genes involved in liver regeneration. However, after the PHx (4 h or 48 h post-surgery), differences in gene expression between CT-O and HF-O were suppressed, as well as in microRNA expression in the liver. Furthermore, both CT-O and HF-O presented a similar regenerative capacity of the liver within 48 h after PHx. Our results suggest that survival and regenerative mechanisms induced by the partial hepatectomy may overcome the epigenetic changes in the liver of offspring programmed by maternal obesity.
Collapse
Affiliation(s)
- Lais A. de Paula Simino
- Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas—UNICAMP, Limeira, Brazil
| | - Marina Figueiredo Fontana
- Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas—UNICAMP, Limeira, Brazil
| | - Thais de Fante
- Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas—UNICAMP, Limeira, Brazil
| | - Carolina Panzarin
- Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas—UNICAMP, Limeira, Brazil
| | | | - Marciane Milanski
- Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas—UNICAMP, Limeira, Brazil
| | - Marcio Alberto Torsoni
- Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas—UNICAMP, Limeira, Brazil
| | - Mina Desai
- The Lundquist Institute and David Geffen School of Medicine at Harbor-UCLA Medical Center, University of California, Los Angeles, Los Angeles, CA, United States
| | - Michael G. Ross
- The Lundquist Institute and David Geffen School of Medicine at Harbor-UCLA Medical Center, University of California, Los Angeles, Los Angeles, CA, United States
| | - Adriana Souza Torsoni
- Laboratory of Metabolic Disorders, School of Applied Sciences, University of Campinas—UNICAMP, Limeira, Brazil
- *Correspondence: Adriana Souza Torsoni,
| |
Collapse
|
24
|
Ko S, Kim M, Molina L, Sirica AE, Monga SP. YAP1 activation and Hippo pathway signaling in the pathogenesis and treatment of intrahepatic cholangiocarcinoma. Adv Cancer Res 2022; 156:283-317. [PMID: 35961703 PMCID: PMC9972177 DOI: 10.1016/bs.acr.2022.02.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Intrahepatic cholangiocarcinoma (iCCA), the second most common primary liver cancer, is a highly lethal epithelial cell malignancy exhibiting features of cholangiocyte differentiation. iCCAs can potentially develop from multiple cell types of origin within liver, including immature or mature cholangiocytes, hepatic stem cells/progenitor cells, and from transdifferentiation of hepatocytes. Understanding the molecular mechanisms and genetic drivers that diversely drive specific cell lineage pathways leading to iCCA has important biological and clinical implications. In this context, activation of the YAP1-TEAD dependent transcription, driven by Hippo-dependent or -independent diverse mechanisms that lead to the stabilization of YAP1 is crucially important to biliary fate commitment in hepatobiliary cancer. In preclinical models, YAP1 activation in hepatocytes or cholangiocytes is sufficient to drive their malignant transformation into iCCA. Moreover, nuclear YAP1/TAZ is highly prevalent in human iCCA irrespective of the varied etiology, and significantly correlates with poor prognosis in iCCA patients. Based on the ubiquitous expression and diverse physiologic roles for YAP1/TAZ in the liver, recent studies have further revealed distinct functions of active YAP1/TAZ in regulating tumor metabolism, as well as the tumor immune microenvironment. In the current review, we discuss our current understanding of the various roles of the Hippo-YAP1 signaling in iCCA pathogenesis, with a specific focus on the roles played by the Hippo-YAP1 pathway in modulating biliary commitment and oncogenicity, iCCA metabolism, and immune microenvironment. We also discuss the therapeutic potential of targeting the YAP1/TAZ-TEAD transcriptional machinery in iCCA, its current limitations, and what future studies are needed to facilitate clinical translation.
Collapse
Affiliation(s)
- Sungjin Ko
- Division of Experimental Pathology, Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States; Pittsburgh Liver Research Center, Pittsburgh, PA, United States.
| | - Minwook Kim
- Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Laura Molina
- Division of Experimental Pathology, Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States; Pittsburgh Liver Research Center, Pittsburgh, PA, United States
| | - Alphonse E Sirica
- Department of Pathology, Virginia Commonwealth University School of Medicine, Richmond, VA, United States
| | - Satdarshan P Monga
- Division of Experimental Pathology, Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States; Pittsburgh Liver Research Center, Pittsburgh, PA, United States; Division of Gastroenterology, Hepatology, and Nutrition, University of Pittsburgh and UPMC, Pittsburgh, PA, United States.
| |
Collapse
|
25
|
Ulhaq ZS, Tse WKF. A Brief Analysis of Proteomic Profile Changes during Zebrafish Regeneration. Biomolecules 2021; 12:biom12010035. [PMID: 35053182 PMCID: PMC8773715 DOI: 10.3390/biom12010035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 12/21/2021] [Accepted: 12/21/2021] [Indexed: 11/16/2022] Open
Abstract
Unlike mammals, zebrafish are capable to regenerate many of their organs, however, the response of tissue damage varies across tissues. Understanding the molecular mechanism behind the robust regenerative capacity in a model organism may help to identify and develop novel treatment strategies for mammals (including humans). Hence, we systematically analyzed the current literature on the proteome profile collected from different regenerated zebrafish tissues. Our analyses underlining that several proteins and protein families responsible as a component of cytoskeleton and structure, protein synthesis and degradation, cell cycle control, and energy metabolism were frequently identified. Moreover, target proteins responsible for the initiation of the regeneration process, such as inflammation and immune response were less frequently detected. This highlights the limitation of previous proteomic analysis and suggested a more sensitive modern proteomics analysis is needed to unfold the mechanism. This brief report provides a list of target proteins with predicted functions that could be useful for further biological studies.
Collapse
Affiliation(s)
- Zulvikar Syambani Ulhaq
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Maulana Malik Ibrahim State Islamic University of Malang, Batu 65144, Indonesia;
- National Research and Innovation Agency, Central Jakarta 10340, Indonesia
| | - William Ka Fai Tse
- Laboratory of Developmental Disorders and Toxicology, Center for Promotion of International Education and Research, Faculty of Agriculture, Kyushu University, Fukuoka 819-0395, Japan
- Correspondence:
| |
Collapse
|
26
|
Liang R, Lin YH, Zhu H. Genetic and Cellular Contributions to Liver Regeneration. Cold Spring Harb Perspect Biol 2021; 14:a040832. [PMID: 34750173 PMCID: PMC9438780 DOI: 10.1101/cshperspect.a040832] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The regenerative capabilities of the liver represent a paradigm for understanding tissue repair in solid organs. Regeneration after partial hepatectomy in rodent models is well understood, while regeneration in the context of clinically relevant chronic injuries is less studied. Given the growing incidence of fatty liver disease, cirrhosis, and liver cancer, interest in liver regeneration is increasing. Here, we will review the principles, genetics, and cell biology underlying liver regeneration, as well as new approaches being used to study heterogeneity in liver tissue maintenance and repair.
Collapse
Affiliation(s)
- Roger Liang
- Children's Research Institute, Departments of Pediatrics and Internal Medicine, Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Yu-Hsuan Lin
- Children's Research Institute, Departments of Pediatrics and Internal Medicine, Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Hao Zhu
- Children's Research Institute, Departments of Pediatrics and Internal Medicine, Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| |
Collapse
|
27
|
Vulf M, Shunkina D, Komar A, Bograya M, Zatolokin P, Kirienkova E, Gazatova N, Kozlov I, Litvinova L. Analysis of miRNAs Profiles in Serum of Patients With Steatosis and Steatohepatitis. Front Cell Dev Biol 2021; 9:736677. [PMID: 34568346 PMCID: PMC8458751 DOI: 10.3389/fcell.2021.736677] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 08/10/2021] [Indexed: 12/17/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is emerging as one of the most common chronic liver diseases worldwide, affecting 25% of the world population. In recent years, there has been increasing evidence for the involvement of microRNAs in the epigenetic regulation of genes taking part in the development of steatosis and steatohepatitis—two main stages of NAFLD pathogenesis. In the present study, miRNA profiles were studied in groups of patients with steatosis and steatohepatitis to compare the characteristics of RNA-dependent epigenetic regulation of the stages of NAFLD development. According to the results of miRNA screening, 23 miRNAs were differentially expressed serum in a group of patients with steatohepatitis and 2 in a group of patients with steatosis. MiR-195-5p and miR-16-5p are common differentially expressed miRNAs for both steatosis and steatohepatitis. We analyzed the obtained results: the search for target genes for the differentially expressed miRNAs in our study and the subsequent gene set enrichment analysis performed on KEGG and REACTOME databases revealed which metabolic pathways undergo changes in RNA-dependent epigenetic regulation in steatosis and steatohepatitis. New findings within the framework of this study are the dysregulation of neurohumoral pathways in the pathogenesis of NAFLD as an object of changes in RNA-dependent epigenetic regulation. The miRNAs differentially expressed in our study were found to target 7% of genes in the classic pathogenesis of NAFLD in the group of patients with steatosis and 50% in the group of patients with steatohepatitis. The effects of these microRNAs on genes for the pathogenesis of NAFLD were analyzed in detail. MiR-374a-5p, miR-1-3p and miR-23a-3p do not target genes directly involved in the pathogenesis of NAFLD. The differentially expressed miRNAs found in this study target genes largely responsible for mitochondrial function. The role of miR-423-5p, miR-143-5p and miR-200c-3 in regulating apoptotic processes in the liver and hepatocarcinogenesis is of interest for future experimental studies. These miR-374a, miR-143, miR-1, miR-23a, and miR-423 have potential for steatohepatitis diagnosis and are poorly studied in the context of NAFLD. Thus, this work opens up prospects for further studies of microRNAs as diagnostic and therapeutic biomarkers for NAFLD.
Collapse
Affiliation(s)
- Maria Vulf
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, Kaliningrad, Russia
| | - Daria Shunkina
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, Kaliningrad, Russia
| | - Aleksandra Komar
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, Kaliningrad, Russia
| | - Maria Bograya
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, Kaliningrad, Russia
| | - Pavel Zatolokin
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, Kaliningrad, Russia
| | - Elena Kirienkova
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, Kaliningrad, Russia
| | - Natalia Gazatova
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, Kaliningrad, Russia
| | - Ivan Kozlov
- Department of Organization and Management in the Sphere of Circulation of Medicines, Institute of Postgraduate Education, I.M. Sechenov Federal State Autonomous Educational University of Higher Education-First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia
| | - Larisa Litvinova
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, Kaliningrad, Russia
| |
Collapse
|
28
|
Zhang L, Ma XJN, Fei YY, Han HT, Xu J, Cheng L, Li X. Stem cell therapy in liver regeneration: Focus on mesenchymal stem cells and induced pluripotent stem cells. Pharmacol Ther 2021; 232:108004. [PMID: 34597754 DOI: 10.1016/j.pharmthera.2021.108004] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 08/11/2021] [Accepted: 09/23/2021] [Indexed: 02/07/2023]
Abstract
The liver has the ability to repair itself after injury; however, a variety of pathological changes in the liver can affect its ability to regenerate, and this could lead to liver failure. Mesenchymal stem cells (MSCs) are considered a good source of cells for regenerative medicine, as they regulate liver regeneration through different mechanisms, and their efficacy has been demonstrated by many animal experiments and clinical studies. Induced pluripotent stem cells, another good source of MSCs, have also made great progress in the establishment of organoids, such as liver disease models, and in drug screening. Owing to the recent developments in MSCs and induced pluripotent stem cells, combined with emerging technologies including graphene, nano-biomaterials, and gene editing, precision medicine and individualized clinical treatment may be realized in the near future.
Collapse
Affiliation(s)
- Lu Zhang
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, PR China; Key Laboratory Biotherapy and Regenerative Medicine of Gansu Province, Lanzhou 730000, PR China; The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, PR China
| | - Xiao-Jing-Nan Ma
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, PR China
| | - Yuan-Yuan Fei
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, PR China; Key Laboratory Biotherapy and Regenerative Medicine of Gansu Province, Lanzhou 730000, PR China
| | - Heng-Tong Han
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, PR China
| | - Jun Xu
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, PR China
| | - Lu Cheng
- Key Laboratory Biotherapy and Regenerative Medicine of Gansu Province, Lanzhou 730000, PR China
| | - Xun Li
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, PR China; Key Laboratory Biotherapy and Regenerative Medicine of Gansu Province, Lanzhou 730000, PR China; Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, Lanzhou 730000, PR China; Hepatopancreatobiliary Surgery Institute of Gansu Province, Lanzhou 730000, PR China; The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, PR China.
| |
Collapse
|
29
|
Ferroptosis Meets Cell-Cell Contacts. Cells 2021; 10:cells10092462. [PMID: 34572111 PMCID: PMC8471828 DOI: 10.3390/cells10092462] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 09/07/2021] [Accepted: 09/13/2021] [Indexed: 12/15/2022] Open
Abstract
Ferroptosis is a regulated form of cell death characterized by iron dependency and increased lipid peroxidation. Initially assumed to be selectively induced in tumour cells, there is increasing evidence that ferroptosis plays an important role in pathophysiology and numerous cell types and tissues. Deregulated ferroptosis has been linked to human diseases, such as neurodegenerative diseases, cardiovascular disorders, and cancer. Along these lines, ferroptosis is a promising pathway to overcoming therapy resistance of cancer cells. It is therefore of utmost importance to understand the cellular signalling pathways and the molecular mechanisms underlying ferroptosis regulation, including context-specific effects mediated by the neighbouring cells through cell–cell contacts. Here, we give an overview on the molecular events and machinery linked to ferroptosis induction and commitment. We further summarize and discuss current knowledge about the role of cell–cell contacts, which differ in ferroptosis regulation between normal somatic cells and cancer cells. We present emerging concepts on the underlying mechanisms, address open questions, and discuss the possible impact of cell–cell contacts on exploiting ferroptosis in cancer therapy.
Collapse
|
30
|
Lv D, Chen L, Du L, Zhou L, Tang H. Emerging Regulatory Mechanisms Involved in Liver Cancer Stem Cell Properties in Hepatocellular Carcinoma. Front Cell Dev Biol 2021; 9:691410. [PMID: 34368140 PMCID: PMC8339910 DOI: 10.3389/fcell.2021.691410] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 07/01/2021] [Indexed: 02/05/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the predominant form of primary liver cancer and one of the leading causes of cancer-related deaths worldwide. A growing body of evidence supports the hypothesis that HCC is driven by a population of cells called liver cancer stem cells (LCSCs). LCSCs have been proposed to contribute to malignant HCC progression, including promoting tumor occurrence and growth, mediating tumor metastasis, and treatment resistance, but the regulatory mechanism of LCSCs in HCC remains unclear. Understanding the signaling pathways responsible for LCSC maintenance and survival may provide opportunities to improve patient outcomes. Here, we review the current literature about the origin of LCSCs and the niche composition, describe the current evidence of signaling pathways that mediate LCSC stemness, then highlight several mechanisms that modulate LCSC properties in HCC progression, and finally, summarize the new developments in therapeutic strategies targeting LCSCs markers and regulatory pathways.
Collapse
Affiliation(s)
- Duoduo Lv
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
| | - Liyu Chen
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
| | - Lingyao Du
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
| | - Lingyun Zhou
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
| | - Hong Tang
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China.,State Key Laboratory of Biotherapy and Center of Infectious Diseases, Division of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
31
|
Brücher VC, Egbring C, Plagemann T, Nedvetsky PI, Höffken V, Pavenstädt H, Eter N, Kremerskothen J, Heiduschka P. Lack of WWC2 Protein Leads to Aberrant Angiogenesis in Postnatal Mice. Int J Mol Sci 2021; 22:5321. [PMID: 34070186 PMCID: PMC8158494 DOI: 10.3390/ijms22105321] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 05/11/2021] [Accepted: 05/13/2021] [Indexed: 01/03/2023] Open
Abstract
The WWC protein family is an upstream regulator of the Hippo signalling pathway that is involved in many cellular processes. We examined the effect of an endothelium-specific WWC1 and/or WWC2 knock-out on ocular angiogenesis. Knock-outs were induced in C57BL/6 mice at the age of one day (P1) and evaluated at P6 (postnatal mice) or induced at the age of five weeks and evaluated at three months of age (adult mice). We analysed morphology of retinal vasculature in retinal flat mounts. In addition, in vivo imaging and functional testing by electroretinography were performed in adult mice. Adult WWC1/2 double knock-out mice differed neither functionally nor morphologically from the control group. In contrast, the retinas of the postnatal WWC knock-out mice showed a hyperproliferative phenotype with significantly enlarged areas of sprouting angiogenesis and a higher number of tip cells. The branching and end points in the peripheral plexus were significantly increased compared to the control group. The deletion of the WWC2 gene was decisive for these effects; while knocking out WWC1 showed no significant differences. The results hint strongly that WWC2 is an essential regulator of ocular angiogenesis in mice. As an activator of the Hippo signalling pathway, it prevents excessive proliferation during physiological angiogenesis. In adult animals, WWC proteins do not seem to be important for the maintenance of the mature vascular plexus.
Collapse
Affiliation(s)
- Viktoria Constanze Brücher
- Department of Ophthalmology, University of Münster Medical School, 48149 Münster, Germany; (V.C.B.); (C.E.); (T.P.); (N.E.)
| | - Charlotte Egbring
- Department of Ophthalmology, University of Münster Medical School, 48149 Münster, Germany; (V.C.B.); (C.E.); (T.P.); (N.E.)
| | - Tanja Plagemann
- Department of Ophthalmology, University of Münster Medical School, 48149 Münster, Germany; (V.C.B.); (C.E.); (T.P.); (N.E.)
- Department of Nephrology, Internal Medicine D, Hypertension and Rheumatology, University of Münster Medical School, 48149 Münster, Germany; (P.I.N.); (H.P.); (J.K.)
| | - Pavel I. Nedvetsky
- Department of Nephrology, Internal Medicine D, Hypertension and Rheumatology, University of Münster Medical School, 48149 Münster, Germany; (P.I.N.); (H.P.); (J.K.)
| | - Verena Höffken
- Medical Cell Biology, Medical Clinic D, University of Münster Medical School, 48149 Münster, Germany;
| | - Hermann Pavenstädt
- Department of Nephrology, Internal Medicine D, Hypertension and Rheumatology, University of Münster Medical School, 48149 Münster, Germany; (P.I.N.); (H.P.); (J.K.)
| | - Nicole Eter
- Department of Ophthalmology, University of Münster Medical School, 48149 Münster, Germany; (V.C.B.); (C.E.); (T.P.); (N.E.)
| | - Joachim Kremerskothen
- Department of Nephrology, Internal Medicine D, Hypertension and Rheumatology, University of Münster Medical School, 48149 Münster, Germany; (P.I.N.); (H.P.); (J.K.)
| | - Peter Heiduschka
- Department of Ophthalmology, University of Münster Medical School, 48149 Münster, Germany; (V.C.B.); (C.E.); (T.P.); (N.E.)
| |
Collapse
|